Network


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

Hotspot


Dive into the research topics where David L. Sloman is active.

Publication


Featured researches published by David L. Sloman.


Bioorganic & Medicinal Chemistry Letters | 2008

Exploration of the internal cavity of histone deacetylase (HDAC) with selective HDAC1/HDAC2 inhibitors (SHI-1:2)

Joey L. Methot; Prasun K. Chakravarty; Melissa Chenard; Joshua Close; Jonathan C. Cruz; William K. Dahlberg; Judith C. Fleming; Christopher Hamblett; Julie E. Hamill; Paul Harrington; Andreas Harsch; Richard Heidebrecht; Bethany Hughes; Joon Jung; Candia M. Kenific; Astrid M. Kral; Peter T. Meinke; Richard E. Middleton; Nicole Ozerova; David L. Sloman; Matthew G. Stanton; Alexander A. Szewczak; Sriram Tyagarajan; David J. Witter; J. Paul Secrist; Thomas A. Miller

We report herein the initial exploration of novel selective HDAC1/HDAC2 inhibitors (SHI-1:2). Optimized SHI-1:2 structures exhibit enhanced intrinsic activity against HDAC1 and HDAC2, and are greater than 100-fold selective versus other HDACs, including HDAC3. Based on the SAR of these agents and our current understanding of the HDAC active site, we postulate that the SHI-1:2 extend the existing HDAC inhibitor pharmacophore to include an internal binding domain.


Natural Product Reports | 2013

Polycyclic xanthone natural products: structure, biological activity and chemical synthesis

Dana K. Winter; David L. Sloman; John A. Porco

Polycyclic xanthone natural products are a family of polyketides which are characterized by highly oxygenated, angular hexacyclic frameworks. In the last decade, this novel class of molecules has attracted noticeable attention from the synthetic and biological communities due to emerging reports of their potential use as antitumour agents. The aim of this article is to highlight the most recent developments of this subset of the xanthone family by detailing the innate challenges of the construction of this class of natural products, new synthetic approaches, and pharmacological data.


Bioorganic & Medicinal Chemistry Letters | 2010

Fluorinated piperidine acetic acids as γ-secretase modulators

Matthew G. Stanton; Jed L. Hubbs; David L. Sloman; Christopher Hamblett; Paula Andrade; Minilik Angagaw; Grace Bi; Regina M. Black; Jamie L. Crispino; Jonathan C. Cruz; Eric Fan; Georgia Farris; Bethany Hughes; Candia M. Kenific; Richard E. Middleton; George Nikov; Peter Sajonz; Sanjiv Shah; Nirah H. Shomer; Alexander A. Szewczak; Flobert Tanga; Matthew T. Tudge; Mark S. Shearman; Benito Munoz

We report herein a novel series of difluoropiperidine acetic acids as modulators of gamma-secretase. Synthesis of 2-aryl-3,3-difluoropiperidine analogs was facilitated by a unique and selective beta-difluorination with Selectfluor. Compounds 1f and 2c were selected for in vivo assessment and demonstrated selective lowering of Abeta42 in a genetically engineered mouse model of APP processing. Moreover, in a 7-day safety study, rats treated orally with compound 1f (250mg/kg per day, AUC(0-24)=2100microMh) did not exhibit Notch-related effects.


Bioorganic & Medicinal Chemistry Letters | 2016

Optimization of microtubule affinity regulating kinase (MARK) inhibitors with improved physical properties.

David L. Sloman; Njamkou Noucti; Michael D. Altman; Dapeng Chen; Andrea C. Mislak; Alexander A. Szewczak; Mansuo L. Hayashi; Lee Warren; Tammy Dellovade; Zhenhua Wu; Jacob Marcus; Deborah Walker; Hua-Poo Su; Suzanne C. Edavettal; Sanjeev Munshi; Michael Hutton; Hugh Nuthall; Matthew G. Stanton

Inhibition of microtubule affinity regulating kinase (MARK) represents a potentially attractive means of arresting neurofibrillary tangle pathology in Alzheimers disease. This manuscript outlines efforts to optimize a pyrazolopyrimidine series of MARK inhibitors by focusing on improvements in potency, physical properties and attributes amenable to CNS penetration. A unique cylcyclohexyldiamine scaffold was identified that led to remarkable improvements in potency, opening up opportunities to reduce MW, Pgp efflux and improve pharmacokinetic properties while also conferring improved solubility.


Cancer Research | 2010

Abstract 5433: Prolonged histone hyperacetylation with a novel class of HDAC1/2 selective inhibitors

Joey L. Methot; Melissa Chenard; Close Joshua; Cruz Jonathan; William K. Dahlberg; Judith C. Fleming; Chris Hamblett; Hamill Julie; Paul Harrington; Andreas Harsch; Richard Heidebrecht; Bethany Hughes; Joon Jung; Candia M. Kenific; Astrid M. Kral; Richard E. Middleton; Nicole Ozerova; David L. Sloman; Matthew G. Stanton; Alexander A. Szewczak; Kevin J. Wilson; David J. Witter; Paul Secrist; Thomas A. Miller

The histone deacetylase (HDAC) metalloenzymes are intricately involved in gene expression through epigenetic regulation of histone acetylation. They also regulate the acetylation status of numerous non-histone proteins such as transcription factors p53, STAT1 and NF-κB as well as α-tubulin, Hsp90 and Ku70. Of the eleven zinc-dependent HDAC enzymes identified, HDACs 1 and 2 appear to be most critical in oncogenesis and tumor maintenance. They are overexpressed in many human cancers and RNAi knockdown leads to increased apoptosis. We recently disclosed a family of novel HDAC1/HDAC2-selective biaryl inhibitors. In this presentation, we will describe unique features of these biaryl inhibitors that contribute to improved preclinical efficacy and tolerability. Desirable HDAC inhibitor properties identified from preclinical experience with Zolinza™ include subtype selectivity toward HDAC1/HDAC2 and prolonged target inhibition. Compelling in vitro and in vivo data indicates that solid tumor cell lines are most sensitive to HDAC inhibition under continuous exposure rather than intermittent exposure. These biaryl inhibitors exhibit extended target engagement in vivo, and are well tolerated in nude mice. Unlike other known HDAC inhibitors, these compounds exhibit a delay in and prolongation of histone hyperacetylation in nude mice bearing HCT116 tumors, extending beyond plasma clearance of the drug. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 5433.


Archive | 2014

Novel compounds that are erk inhibitors

Kevin J. Wilson; David J. Witter; Matthew H. Daniels; Angie R. Angeles; Phieng Siliphaivanh; David L. Sloman; Brendan M. O'boyle; Danielle Falcone; Catherine White; Ron Ferguson; Wei Zhou; Kathryn Lipford; Umar Faruk Mansoor; Salem Fevrier; Xianhai Huang; Ravi Kurukulasuriya; Judson E. Richard; Shuyi Tang; Christopher W. Boyce; Joseph A. Kozlowski; Raman K. Bakshi; Ganesh Babu Karunakaran


Bioorganic & Medicinal Chemistry Letters | 2007

The discovery of 6-amino nicotinamides as potent and selective histone deacetylase inhibitors.

Christopher Hamblett; Joey L. Methot; Dawn M. Mampreian; David L. Sloman; Matthew G. Stanton; Astrid M. Kral; Judith C. Fleming; Jonathan C. Cruz; Melissa Chenard; Nicole Ozerova; Anna M. Hitz; Hongmei Wang; Sujal V. Deshmukh; Naim Nazef; Andreas Harsch; Bethany Hughes; William K. Dahlberg; Alex A. Szewczak; Richard E. Middleton; Ralph T. Mosley; J. Paul Secrist; Thomas A. Miller


Archive | 2009

Compounds that are erk inhibitors

Kevin J. Wilson; David J. Witter; Phieng Siliphaivanh; Kathryn Lipford; David L. Sloman; Danielle Falcone; Brendan M. O'boyle; Umar Faruk Mansoor; Jongwon Lim; Joey L. Methot; Christopher W. Boyce; Lei Chen; Matthew H. Daniels; Salem Fevrier; Xianhai Huang; Ravi Kurukulasuriya; Ling Tong; Wei Zhou; Joseph A. Kozlowski; Milana Maletic; Bidhan A. Shinkre; Jayanth Thiruvellore Thatai; Raman K. Bakshi; Ganesh Babu Karunakaran


Archive | 2006

Histone deacetylase inhibitors with aryl-pyrazolyl motifs

Joshua Close; Richard Heidebrecht; Solomon Kattar; Thomas A. Miller; David L. Sloman; Matthew G. Stanton; Paul Tempest; David J. Witter


Archive | 2006

Spirocyclic compounds as hdac inhibitors

Scott C. Berk; Joshua Close; Christopher Hamblett; Richard Heidebrecht; Solomon Kattar; Laura T. Kliman; Dawn M. Mampreian; Joey L. Methot; Thomas A. Miller; David L. Sloman; Matthew G. Stanton; Paul Tempest; Anna A. Zabierek

Researchain Logo
Decentralizing Knowledge