Network


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

Hotspot


Dive into the research topics where Charles Kreiman is active.

Publication


Featured researches published by Charles Kreiman.


Journal of Medicinal Chemistry | 2012

Design and preparation of a potent series of hydroxyethylamine containing β-secretase inhibitors that demonstrate robust reduction of central β-amyloid.

Matthew Weiss; Toni Williamson; Safura Babu-Khan; Michael D. Bartberger; James Brown; Kui Chen; Yuan Cheng; Martin Citron; Michael Croghan; Thomas Dineen; Joel Esmay; Russell Graceffa; Scott Harried; Dean Hickman; Stephen A. Hitchcock; Daniel B. Horne; Hongbing Huang; Ronke Imbeah-Ampiah; Ted Judd; Matthew R. Kaller; Charles Kreiman; Daniel S. La; Vivian Li; Patricia Lopez; Steven W. Louie; Holger Monenschein; Thomas Nguyen; Lewis D. Pennington; Claire Rattan; Tisha San Miguel

A series of potent hydroxyethyl amine (HEA) derived inhibitors of β-site APP cleaving enzyme (BACE1) was optimized to address suboptimal pharmacokinetics and poor CNS partitioning. This work identified a series of benzodioxolane analogues that possessed improved metabolic stability and increased oral bioavailability. Subsequent efforts focused on improving CNS exposure by limiting susceptibility to Pgp-mediated efflux and identified an inhibitor which demonstrated robust and sustained reduction of CNS β-amyloid (Aβ) in Sprague-Dawley rats following oral administration.


Journal of Medicinal Chemistry | 2012

Design and synthesis of potent, orally efficacious hydroxyethylamine derived β-site amyloid precursor protein cleaving enzyme (BACE1) inhibitors.

Thomas Dineen; Matthew Weiss; Toni Williamson; Paul D. Acton; Safura Babu-Khan; Michael D. Bartberger; James Brown; Kui Chen; Yuan Cheng; Martin Citron; Michael Croghan; Robert Dunn; Joel Esmay; Russell Graceffa; Scott Harried; Dean Hickman; Stephen A. Hitchcock; Daniel B. Horne; Hongbing Huang; Ronke Imbeah-Ampiah; Ted Judd; Matthew R. Kaller; Charles Kreiman; Daniel S. La; Vivian Li; Patricia Lopez; Steven W. Louie; Holger Monenschein; Thomas Nguyen; Lewis D. Pennington

We have previously shown that hydroxyethylamines can be potent inhibitors of the BACE1 enzyme and that the generation of BACE1 inhibitors with CYP 3A4 inhibitory activities in this scaffold affords compounds (e.g., 1) with sufficient bioavailability and pharmacokinetic profiles to reduce central amyloid-β peptide (Aβ) levels in wild-type rats following oral dosing. In this article, we describe further modifications of the P1-phenyl ring of the hydroxyethylamine series to afford potent, dual BACE1/CYP 3A4 inhibitors which demonstrate improved penetration into the CNS. Several of these compounds caused robust reduction of Aβ levels in rat CSF and brain following oral dosing, and compound 37 exhibited an improved cardiovascular safety profile relative to 1.


Bioorganic & Medicinal Chemistry Letters | 2015

Development of 2-aminooxazoline 3-azaxanthenes as orally efficacious β-secretase inhibitors for the potential treatment of Alzheimer's disease.

Jian Jeffrey Chen; Qingyian Liu; Chester Chenguang Yuan; Vijay Keshav Gore; Patricia Lopez; Vu Van Ma; Albert Amegadzie; Wenyuan Qian; Ted Judd; Ana Elena Minatti; James Brown; Yuan Cheng; May Xue; Wenge Zhong; Thomas Dineen; Oleg Epstein; Jason Brooks Human; Charles Kreiman; Isaac E. Marx; Matthew Weiss; Stephen A. Hitchcock; Timothy Powers; Kui Chen; Paul H. Wen; Douglas A. Whittington; Alan C. Cheng; Michael D. Bartberger; Dean Hickman; Jonathan A. Werner; Hugo M. Vargas

The β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1) is one of the most hotly pursued targets for the treatment of Alzheimers disease. We used a structure- and property-based drug design approach to identify 2-aminooxazoline 3-azaxanthenes as potent BACE1 inhibitors which significantly reduced CSF and brain Aβ levels in a rat pharmacodynamic model. Compared to the initial lead 2, compound 28 exhibited reduced potential for QTc prolongation in a non-human primate cardiovascular safety model.


Journal of Medicinal Chemistry | 2017

Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics While Mitigating Metabolic Liabilities

Matthew Weiss; Thomas Dineen; Isaac E. Marx; Steven Altmann; Alessandro Boezio; Howard Bregman; Margaret Y. Chu-Moyer; Erin F. DiMauro; Elma Feric Bojic; Robert S. Foti; Hua Gao; Russell Graceffa; Hakan Gunaydin; Angel Guzman-Perez; Hongbing Huang; Liyue Huang; Michael Jarosh; Thomas Kornecook; Charles Kreiman; Joseph Ligutti; Daniel S. La; Min-Hwa Jasmine Lin; Dong Liu; Bryan D. Moyer; Hanh Nho Nguyen; Emily A. Peterson; Paul Rose; Kristin Taborn; Beth D. Youngblood; Violeta Yu

Several reports have recently emerged regarding the identification of heteroarylsulfonamides as NaV1.7 inhibitors that demonstrate high levels of selectivity over other NaV isoforms. The optimization of a series of internal NaV1.7 leads that address a number of metabolic liabilities including bioactivation, PXR activation, as well as CYP3A4 induction and inhibition led to the identification of potent and selective inhibitors that demonstrated favorable pharmacokinetic profiles and were devoid of the aforementioned liabilities. The key to achieving this within a series prone to transporter-mediated clearance was the identification of a small range of optimal cLogD values and the discovery of subtle PXR SAR that was not lipophilicity dependent. This enabled the identification of compound 20, which was advanced into a target engagement pharmacodynamic model where it exhibited robust reversal of histamine-induced scratching bouts in mice.


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.


Bioorganic & Medicinal Chemistry Letters | 2017

The discovery of benzoxazine sulfonamide inhibitors of NaV1.7: Tools that bridge efficacy and target engagement

Daniel S. La; Emily A. Peterson; Christiane Bode; Alessandro Boezio; Howard Bregman; Margaret Yuhua Chu-Moyer; James R. Coats; Erin F. DiMauro; Thomas Dineen; Bingfan Du; Hua Gao; Russell Graceffa; Hakan Gunaydin; Angel Guzman-Perez; Robert T. Fremeau; Xin Huang; Christopher P. Ilch; Thomas Kornecook; Charles Kreiman; Joseph Ligutti; Min-Hwa Jasmine Lin; Jeff S. McDermott; Isaac E. Marx; David J. Matson; Bryan D. Moyer; Hanh Nho Nguyen; Kristin Taborn; Violeta Yu; Matthew Weiss

The voltage-gated sodium channel NaV1.7 has received much attention from the scientific community due to compelling human genetic data linking gain- and loss-of-function mutations to pain phenotypes. Despite this genetic validation of NaV1.7 as a target for pain, high quality pharmacological tools facilitate further understanding of target biology, establishment of target coverage requirements and subsequent progression into the clinic. Within the sulfonamide class of inhibitors, reduced potency on rat NaV1.7 versus human NaV1.7 was observed, rendering in vivo rat pharmacology studies challenging. Herein, we report the discovery and optimization of novel benzoxazine sulfonamide inhibitors of human, rat and mouse NaV1.7 which enabled pharmacological assessment in traditional behavioral rodent models of pain and in turn, established a connection between formalin-induced pain and histamine-induced pruritus in mice. The latter represents a simple and efficient means of measuring target engagement.


ACS Medicinal Chemistry Letters | 2017

Correction to “Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics to Enable in Vivo Target Engagement”

Isaac E. Marx; Thomas Dineen; Jessica Able; Christiane Bode; Howard Bregman; Margaret Y. Chu-Moyer; Erin F. DiMauro; Bingfan Du; Robert S. Foti; Robert T. Fremeau; Hua Gao; Hakan Gunaydin; Brian E. Hall; Liyue Huang; Thomas Kornecook; Charles Kreiman; Daniel S. La; Joseph Ligutti; Min-Hwa Jasmine Lin; Dong Liu; Jeff S. McDermott; Bryan D. Moyer; Hanh Nho Nguyen; Emily A. Peterson; Jonathan Roberts; Paul Rose; Jean Wang; Beth D. Youngblood; Violeta Yu; Matthew Weiss

[This corrects the article DOI: 10.1021/acsmedchemlett.6b00243.].


Archive | 2009

Spiro-tetracyclic ring compounds as beta-secretase modulators and methods of use

Yuan Cheng; Ryan White; Albert Amegadzie; James Brown; Alan C. Cheng; Erin F. DiMauro; Thomas Dineen; Oleg Epstein; Vijay Keshav Gore; Jason Brooks Human; Ted Judd; Charles Kreiman; Qingyian Liu; Patricia Lopez; Vu Van Ma; Isaac E. Marx; Ana Elena Minatti; Hanh Nho Nguyen; Nick A. Paras; Vinod F. Patel; Wenyuan Qian; Matthew Weiss; Qiufen Xue; Xiao Mei Zheng; Wenge Zhong


ACS Medicinal Chemistry Letters | 2016

Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics to Enable in Vivo Target Engagement

Isaac E. Marx; Thomas Dineen; Jessica Able; Christiane Bode; Howard Bregman; Margaret Y. Chu-Moyer; Erin F. DiMauro; Bingfan Du; Robert S. Foti; Robert T. Fremeau; Hua Gao; Hakan Gunaydin; Brian E. Hall; Liyue Huang; Thomas Kornecook; Charles Kreiman; Daniel S. La; Joseph Ligutti; Min-Hwa Jasmine Lin; Dong Liu; Jeff S. McDermott; Bryan D. Moyer; Emily A. Peterson; Jonathan Roberts; Paul Rose; Jean Wang; Beth D. Youngblood; Violeta Yu; Matthew Weiss


Archive | 2006

Beta-secretase modulators and methods of use

Brian K. Albrecht; Denise Lyn Andersen; Michael D. Bartberger; James Brown; Ryan Brown; Stuart C. Chaffee; Yuan Cheng; Michael Croghan; Russell Graceffa; Scott Harried; Stephen A. Hitchcock; Randall W. Hungate; Ted Judd; Matthew R. Kaller; Charles Kreiman; Daniel La; Patricia Lopez; Craig E. Masse; Holger Monenschein; Thomas Nguyen; Thomas Nixey; Vinod F. Patel; Lewis D. Pennington; Matthew Weiss; Qiufen Xue; Bryant Yang; Wenge Zhong

Collaboration


Dive into the Charles Kreiman's collaboration.

Researchain Logo
Decentralizing Knowledge