Network


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

Hotspot


Dive into the research topics where Jason D. Katz is active.

Publication


Featured researches published by Jason D. Katz.


Cancer Research | 2010

MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor

Bo-Sheng Pan; Grace K.Y. Chan; Melissa Chenard; An Chi; Lenora Davis; Sujal V. Deshmukh; Jackson B. Gibbs; Susana Gil; Gaozhen Hang; Harold Hatch; James P. Jewell; Ilona Kariv; Jason D. Katz; Kaiko Kunii; Wei Lu; Bart Lutterbach; Cloud P. Paweletz; Xianlu Qu; John F. Reilly; Alexander A. Szewczak; Qinwen Zeng; Nancy E. Kohl; Christopher J. Dinsmore

The receptor tyrosine kinase c-Met is an attractive target for therapeutic blockade in cancer. Here, we describe MK-2461, a novel ATP-competitive multitargeted inhibitor of activated c-Met. MK-2461 inhibited in vitro phosphorylation of a peptide substrate recognized by wild-type or oncogenic c-Met kinases (N1100Y, Y1230C, Y1230H, Y1235D, and M1250T) with IC(50) values of 0.4 to 2.5 nmol/L. In contrast, MK-2461 was several hundredfold less potent as an inhibitor of c-Met autophosphorylation at the kinase activation loop. In tumor cells, MK-2461 effectively suppressed constitutive or ligand-induced phosphorylation of the juxtamembrane domain and COOH-terminal docking site of c-Met, and its downstream signaling to the phosphoinositide 3-kinase-AKT and Ras-extracellular signal-regulated kinase pathways, without inhibiting autophosphorylation of the c-Met activation loop. BIAcore studies indicated 6-fold tighter binding to c-Met when it was phosphorylated, suggesting that MK-2461 binds preferentially to activated c-Met. MK-2461 displayed significant inhibitory activities against fibroblast growth factor receptor (FGFR), platelet-derived growth factor receptor, and other receptor tyrosine kinases. In cell culture, MK-2461 inhibited hepatocyte growth factor/c-Met-dependent mitogenesis, migration, cell scatter, and tubulogenesis. Seven of 10 MK-2461-sensitive tumor cell lines identified from a large panel harbored genomic amplification of MET or FGFR2. In a murine xenograft model of c-Met-dependent gastric cancer, a well-tolerated oral regimen of MK-2461 administered at 100 mg/kg twice daily effectively suppressed c-Met signaling and tumor growth. Similarly, MK-2461 inhibited the growth of tumors formed by s.c. injection of mouse NIH-3T3 cells expressing oncogenic c-Met mutants. Taken together, our findings support further preclinical development of MK-2461 for cancer therapy.


Journal of Medicinal Chemistry | 2011

Discovery of a 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one (MK-2461) inhibitor of c-Met kinase for the treatment of cancer.

Jason D. Katz; James P. Jewell; David J. Guerin; Jongwon Lim; Christopher J. Dinsmore; Sujal V. Deshmukh; Bo-Sheng Pan; C. Gary Marshall; Wei Lu; Michael D. Altman; William K. Dahlberg; Lenora Davis; Danielle Falcone; Ana E. Gabarda; Gaozhen Hang; Harold Hatch; Rachael Holmes; Kaiko Kunii; Kevin J. Lumb; Bart Lutterbach; Robert J. Mathvink; Naim Nazef; Sangita B. Patel; Xianlu Qu; John Reilly; Keith Rickert; Craig Rosenstein; Stephen M. Soisson; Kerrie Spencer; Alexander A. Szewczak

c-Met is a transmembrane tyrosine kinase that mediates activation of several signaling pathways implicated in aggressive cancer phenotypes. In recent years, research into this area has highlighted c-Met as an attractive cancer drug target, triggering a number of approaches to disrupt aberrant c-Met signaling. Screening efforts identified a unique class of 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one kinase inhibitors, exemplified by 1. Subsequent SAR studies led to the development of 81 (MK-2461), a potent inhibitor of c-Met that was efficacious in preclinical animal models of tumor suppression. In addition, biochemical studies and X-ray analysis have revealed that this unique class of kinase inhibitors binds preferentially to the activated (phosphorylated) form of the kinase. This report details the development of 81 and provides a description of its unique biochemical properties.


Journal of Organic Chemistry | 2009

Preparation of a stable trifluoroborate salt for the synthesis of 1-aryl-2,2-difluoro-enolethers and/or 2,2-difluoro-1-aryl-ketones via palladium-mediated cross-coupling.

Jason D. Katz; Blair T. Lapointe; Christopher J. Dinsmore

A bench-stable potassium trifluoroborate enol ether reagent has been prepared. This reagent is suitable for the incorporation of 2,2-difluoroenolethers into aryl and heteroaryl systems via palladium-mediated cross-coupling with suitable halide coupling partners.


Bioorganic & Medicinal Chemistry Letters | 2016

Structure-based design and development of (benz)imidazole pyridones as JAK1-selective kinase inhibitors

Vladimir Simov; Sujal V. Deshmukh; Christopher J. Dinsmore; Fiona Elwood; Rafael Fernandez; Yudith Garcia; Craig R. Gibeau; Hakan Gunaydin; Joon Jung; Jason D. Katz; Brian Kraybill; Blair T. Lapointe; Sangita B. Patel; Tony Siu; Hua Su; Jonathan R. Young

The mammalian Janus Kinases (JAK1, JAK2, JAK3 and TYK2) are intracellular, non-receptor tyrosine kinases whose activities have been associated in the literature and the clinic with a variety of hyperproliferative diseases and immunological disorders. At the onset of the program, it was hypothesized that a JAK1 selective compound over JAK2 could lead to an improved therapeutic index relative to marketed non-selective JAK inhibitors by avoiding the clinical AEs, such as anemia, presumably associated with JAK2 inhibition. During the course of the JAK1 program, a number of diverse chemical scaffolds were identified from both uHTS campaigns and de novo scaffold design. As part of this effort, a (benz)imidazole scaffold evolved via a scaffold-hopping exercise from a mature chemical series. Concurrent crystallography-driven exploration of the ribose pocket and the solvent front led to analogs with optimized kinome and JAK1 selectivities over the JAK2 isoform by targeting several residues unique to JAK1, such as Arg-879 and Glu-966.


Bioorganic & Medicinal Chemistry Letters | 2017

Structure guided design of a series of selective pyrrolopyrimidinone MARK inhibitors

Jason D. Katz; Andrew M. Haidle; Kaleen Konrad Childers; Anna A. Zabierek; James P. Jewell; Yongquan Hou; Michael D. Altman; Alexander A. Szewczak; Dapeng Chen; Andreas Harsch; Mansuo L. Hayashi; Lee Warren; Michael Hutton; Hugh Nuthall; Hua-Poo Su; Sanjeev Munshi; Matt G. Stanton; Ian W. Davies; Ben Munoz; Alan B. Northrup

The initial structure activity relationships around an isoindoline uHTS hit will be described. Information gleaned from ligand co-crystal structures allowed for rapid refinements in both MARK potency and kinase selectivity. These efforts allowed for the identification of a compound with properties suitable for use as an in vitro tool compound for validation studies on MARK as a viable target for Alzheimers disease.


Bioorganic & Medicinal Chemistry Letters | 2017

MARK inhibitors: Declaring a No-Go decision on a chemical series based on extensive DMPK experimentation

Andrew M. Haidle; Kaleen Konrad Childers; Anna A. Zabierek; Jason D. Katz; James P. Jewell; Yongquan Hou; Michael D. Altman; Alexander A. Szewczak; Dapeng Chen; Andreas Harsch; Mansuo L. Hayashi; Lee Warren; Michael Hutton; Hugh Nuthall; Matt G. Stanton; Ian W. Davies; Ben Munoz; Alan B. Northrup

Attempts to optimize pharmacokinetic properties in a promising series of pyrrolopyrimidinone MARK inhibitors for the treatment of Alzheimers disease are described. A focus on physical properties and ligand efficiency while prosecuting this series afforded key tool compounds that revealed a large discrepancy in the rat in vitro-in vivo DMPK (Drug Metabolism/Pharmacokinetics) correlation. These differences prompted an in vivo rat disposition study employing a radiolabeled representative of the series, and the results from this experiment justified the termination of any further optimization efforts.


Archive | 2015

Purine inhibitors of human phosphatidylinositol 3-kinase delta

Abdelghani Abe Achab; Michael D. Altman; Yongqi Deng; Solomon Kattar; Jason D. Katz; Joey L. Methot; Hua Zhou; Meredeth Mcgowan; Matthew Christopher; Yudith Garcia; Neville J. Anthony; Francesc Xavier Fradera Llinas; Liping Yang; Changwei Mu; Xiaona Wang; Feng Shi; Baijun Ye; Sixing Zhang; Xiaoli Zhao; Rong Zhang; Kin Chiu Fong; Xiansheng Leng


Archive | 2009

PYRAZOLO[1,5-A]PYRIDINES AS MARK INHIBITORS

Jason D. Katz; Sandra Lee Knowles; James P. Jewell; David L. Sloman; Matthew G. Stanton; Njamkou Noucti


Archive | 2014

Cycloalkyl nitrile pyrazolo pyridones as janus kinase inhibitors

Matthew Lloyd Childers; Christopher J. Dinsmore; Peter Fuller; David J. Guerin; Jason D. Katz; Qinglin Pu; Mark E. Scott; Christopher F. Thompson; Hongjun Zhang; Danielle Falcone; Luis Torres; Jason Brubaker; Hongbo Zeng; Jiaqiang Cai; Xiaoxing Du; Chonggang Wang; Yunfeng Bai; Norman Kong; Yumei Liu; Zhixiang Zheng


Archive | 2014

Acyclic cyanoethylpyrazolo pyridones as janus kinase inhibitors

Christopher J. Dinsmore; Peter Fuller; David J. Guerin; Jason D. Katz; Christopher F. Thompson; Danielle Falcone; Wei Deng; Luis Torres; Hongbo Zeng; Yunfeng Bai; Jianmin Fu; Norman Kong; Yumei Liu; Zhixiang Zheng; Mark E. Scott

Collaboration


Dive into the Jason D. Katz's collaboration.

Researchain Logo
Decentralizing Knowledge