Network


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

Hotspot


Dive into the research topics where Michael J. Frohn is active.

Publication


Featured researches published by Michael J. Frohn.


Bioorganic & Medicinal Chemistry Letters | 2008

Discovery of novel hydroxy-thiazoles as HIF-α prolyl hydroxylase inhibitors : SAR, synthesis, and modeling evaluation

Christopher M. Tegley; Vellarkad N. Viswanadhan; Kaustav Biswas; Michael J. Frohn; Tanya Peterkin; Catherine H. Chang; Roland W. Bürli; Jennifer Dao; Henrike Veith; Norma Rogers; Sean C. Yoder; Gloria Biddlecome; Philip Tagari; Jennifer R. Allen; Randall W. Hungate

Inhibition of the PHD2 enzyme has been associated with increased red blood cell levels. From a screening hit, a series of novel hydroxyl-thiazoles were developed as potent PHD2 inhibitors.


ACS Medicinal Chemistry Letters | 2011

4-Methoxy-N-[2-(trifluoromethyl)biphenyl-4-ylcarbamoyl]nicotinamide: A Potent and Selective Agonist of S1P1

Lewis D. Pennington; Kelvin Sham; Alexander J. Pickrell; Paul Harrington; Michael J. Frohn; Brian A. Lanman; Anthony B. Reed; Michael Croghan; Matthew R. Lee; Han Xu; Michele McElvain; Yang Xu; Xuxia Zhang; Michael Fiorino; Michelle Horner; Henry Morrison; Heather A. Arnett; Christopher Fotsch; Min Wong; Victor J. Cee

The sphingosine-1-phosphate-1 receptor (S1P1) and its endogenous ligand sphingosine-1-phosphate (S1P) cooperatively regulate lymphocyte trafficking from the lymphatic system. Herein, we disclose 4-methoxy-N-[2-(trifluoromethyl)biphenyl-4-ylcarbamoyl]nicotinamide (8), an uncommon example of a synthetic S1P1 agonist lacking a polar headgroup, which is shown to effect dramatic reduction of circulating lymphocytes (POC = -78%) in rat 24 h after a single oral dose (1 mg/kg). The excellent potency that 8 exhibits toward S1P1 (EC50 = 0.035 μM, 96% efficacy) and the >100-fold selectivity that it displays against receptor subtypes S1P2-5 suggest that it may serve as a valuable tool to understand the clinical relevance of selective S1P1 agonism.


Bioorganic & Medicinal Chemistry | 2014

Synthesis and preliminary biological evaluation of potent and selective 2-(3-alkoxy-1-azetidinyl) quinolines as novel PDE10A inhibitors with improved solubility.

Robert M. Rzasa; Michael J. Frohn; Kristin L. Andrews; Samer Chmait; Ning Chen; Jeffrey Clarine; Carl Davis; Heather Eastwood; Daniel B. Horne; Essa Hu; Adrie D. Jones; Matthew R. Kaller; Roxanne Kunz; Silke Miller; Holger Monenschein; Thomas Nguyen; Alexander J. Pickrell; Amy Porter; Andreas Reichelt; Xiaoning Zhao; James J. S. Treanor; Jennifer R. Allen

We report the discovery of a novel series of 2-(3-alkoxy-1-azetidinyl) quinolines as potent and selective PDE10A inhibitors. Structure-activity studies improved the solubility (pH 7.4) and maintained high PDE10A activity compared to initial lead compound 3, with select compounds demonstrating good oral bioavailability. X-ray crystallographic studies revealed two distinct binding modes to the catalytic site of the PDE10A enzyme. An ex vivo receptor occupancy assay in rats demonstrated that this series of compounds covered the target within the striatum.


Bioorganic & Medicinal Chemistry Letters | 2012

Quinolinone-based agonists of S1P1: Use of a N-scan SAR strategy to optimize in vitro and in vivo activity

Lewis D. Pennington; Michael Croghan; Kelvin Sham; Alexander J. Pickrell; Paul E. Harrington; Michael J. Frohn; Brian A. Lanman; Anthony B. Reed; Matthew R. Lee; Han Xu; Michele McElvain; Yang Xu; Xuxia Zhang; Michael Fiorino; Michelle Horner; Henry Morrison; Heather A. Arnett; Christopher Fotsch; Andrew Tasker; Min Wong; Victor J. Cee

We reveal how a N-scan SAR strategy (systematic substitution of each CH group with a N atom) was employed for quinolinone-based S1P(1) agonist 5 to modulate physicochemical properties and optimize in vitro and in vivo activity. The diaza-analog 17 displays improved potency (hS1P(1) RI; 17: EC(50)=0.020 μM, 120% efficacy; 5: EC(50)=0.070 μM, 110% efficacy) and selectivity (hS1P(3) Ca(2+) flux; 17: EC(50) >25 μM; 5: EC(50)=1.5 μM, 92% efficacy), as well as enhanced pharmacokinetics (17: CL=0.15 L/h/kg, V(dss)=5.1L/kg, T(1/2)=24h, %F=110; 5: CL=0.93L/h/kg, V(dss)=11L/kg, T(1/2)=15 h, %F=60) and pharmacodynamics (17: 1.0mg/kg po, 24h PLC POC=-67%; 5: 3mg/kg po, 24h PLC POC=-51%) in rat.


Cancer Research | 2013

Abstract 711: Small molecule compounds that target cell division cycle 7 (Cdc7) kinase inhibit cell proliferation and tumor growth.

Julie M. Bailis; Li Fang; Jessica Orf; Scott Heller; Tammy L. Bush; Matthew P. Bourbeau; Sonia Escobar; Michael J. Frohn; Paul E. Harrington; Faye Hsieh; Alexander J. Pickrell; Kelvin Sham; Aaron C. Siegmund; Helming Tan; Leeanne Zalameda; John G. Allen; Dineli Wickramasinghe

Cdc7 is an essential, serine/threonine protein kinase that activates the initiation of DNA synthesis at replication origins. Cdc7 also promotes cell cycle checkpoint activation in response to replication stress. As a key regulator of S phase entry and progression, Cdc7 kinase is a potential target for cancer therapy, with a distinct mechanism of action from known drugs that inhibit DNA replication. Following a high throughput screen for inhibitors of Cdc7 kinase activity, we investigated structure-activity relationships of azole-based compounds and optimized the compounds for potency and pharmacokinetic properties. Here we present the characterization of one of these compounds as a potent, selective, bioavailable Cdc7 kinase inhibitor. In cells, Cdc7 inhibition decreases MCM2 phosphorylation and DNA synthesis, causes DNA damage, and slows S phase progression. Cdc7 inhibition also induces chromosome missegregation leading to cell lethality in vitro and tumor growth inhibition in vivo. Cdc7 inhibition provides a new approach to target cancers, either as a single agent or in combination with chemotherapy. Citation Format: Julie Bailis, Li Fang, Jessica Orf, Scott Heller, Tammy Bush, Matthew Bourbeau, Sonia Escobar, Michael Frohn, Paul Harrington, Faye Hsieh, Alexander Pickrell, Kelvin Sham, Aaron Siegmund, Helming Tan, Leeanne Zalameda, John Allen, Dineli Wickramasinghe. Small molecule compounds that target cell division cycle 7 (Cdc7) kinase inhibit cell proliferation and tumor growth. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 711. doi:10.1158/1538-7445.AM2013-711


Archive | 2010

Quinolone based compounds exhibiting, prolyl hydroxylase inhibitory activity, and compositions, and uses thereof

Jennifer R. Allen; Kaustav Biswas; Roland W. Bürli; Jennifer Dao; Michael J. Frohn; Jennifer E. Golden; Randall W. Hungate; Robert J.M. Kurzeja; Stephanie J. Mercede; Kristine M. Muller; Susana C. Neira; Tanya Peterkin; Christopher M. Tegley; Violeta Yu


Archive | 2008

THIENOPYRIDINE AND THIAZOLOPYRIDINE DERIVATIVES THAT INHIBIT PROLYL HYDROXYLASE ACTIVITY

Jennifer R. Allen; Roland W. Bürli; Michael J. Frohn; Randall W. Hungate; Susana C. Neira; Anthony B. Reed


Archive | 2009

S1P1 receptor agonists and use thereof

Victor J. Cee; Michael J. Frohn; Brian A. Lanman; Susana C. Neira; Anthony B. Reed; Kelvin Sham


Archive | 2007

Naphthalenone compounds exhibiting prolyl hydroxylase inhibitory activity, compositions, and uses thereof

Jennifer R. Allen; Kaustav Biswas; Marian C. Bryan; Roland W. Bürli; Guo-Qiang Cao; Michael J. Frohn; Jennifer E. Golden; Stephanie J. Mercede; Susana C. Neira; Tanya Peterkin; Alexander J. Pickrell; Anthony B. Reed; Christopher M. Tegley; Xiang Wang


Archive | 2011

Nitrogen heterocyclic compounds useful as pde10 inhibitors

Jennifer R. Allen; Jian J. Chen; Michael J. Frohn; Essa Hu; Qingyian Liu; Alexander J. Pickrell; Shannon Rumfelt; Robert M. Rzasa; Wenge Zhong

Collaboration


Dive into the Michael J. Frohn's collaboration.

Researchain Logo
Decentralizing Knowledge