Network


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

Hotspot


Dive into the research topics where Andreas Harsch is active.

Publication


Featured researches published by Andreas Harsch.


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.


Bioorganic & Medicinal Chemistry Letters | 2008

SAR profiles of spirocyclic nicotinamide derived selective HDAC1/HDAC2 inhibitors (SHI-1:2)

Joey L. Methot; Christopher Hamblett; Dawn M. Mampreian; Joon Jung; Andreas Harsch; Alexander A. Szewczak; William K. Dahlberg; Richard E. Middleton; Bethany Hughes; Judith C. Fleming; Hongmei Wang; Astrid M. Kral; Nicole Ozerova; Jonathan C. Cruz; Brian B. Haines; Melissa Chenard; Candia M. Kenific; J. Paul Secrist; Thomas A. Miller

A potent family of spirocyclic nicotinyl aminobenzamide selective HDAC1/HDAC2 inhibitors (SHI-1:2) is profiled. The incorporation of a biaryl zinc-binding motif into a nicotinyl scaffold resulted in enhanced potency and selectivity versus HDAC3, but also imparted hERG activity. It was discovered that increasing polar surface area about the spirocycle attenuates this liability. Compound 12 induced a 4-fold increase in acetylated histone H2B in an HCT-116 xenograft model study with acute exposure, and inhibited tumor growth in a 21-day efficacy study with qd dosing.


Bioorganic & Medicinal Chemistry Letters | 2008

Phenylglycine and phenylalanine derivatives as potent and selective HDAC1 inhibitors (SHI-1)

Kevin J. Wilson; David J. Witter; Jonathan Grimm; Phieng Siliphaivanh; Karin M. Otte; Astrid M. Kral; Judith C. Fleming; Andreas Harsch; Julie E. Hamill; Jonathan C. Cruz; Melissa Chenard; Alexander A. Szewczak; Richard E. Middleton; Bethany Hughes; William K. Dahlberg; J. Paul Secrist; Thomas A. Miller

An HTS screening campaign identified a series of low molecular weight phenols that showed excellent selectivity (>100-fold) for HDAC1/HDAC2 over other Class I and Class II HDACs. Evolution and optimization of this HTS hit series provided HDAC1-selective (SHI-1) compounds with excellent anti-proliferative activity and improved physical properties. Dose-dependent efficacy in a mouse HCT116 xenograft model was demonstrated with a phenylglycine SHI-1 analog.


Bioorganic & Medicinal Chemistry Letters | 2009

Parallel medicinal chemistry approaches to selective HDAC1/HDAC2 inhibitor (SHI-1:2) optimization

Solomon Kattar; Laura Surdi; Anna A. Zabierek; Joey L. Methot; Richard E. Middleton; Bethany Hughes; Alexander A. Szewczak; William K. Dahlberg; Astrid M. Kral; Nicole Ozerova; Judith C. Fleming; Hongmei Wang; Paul Secrist; Andreas Harsch; Julie E. Hamill; Jonathan C. Cruz; Candia M. Kenific; Melissa Chenard; Thomas A. Miller; Scott C. Berk; Paul Tempest

The successful application of both solid and solution phase library synthesis, combined with tight integration into the medicinal chemistry effort, resulted in the efficient optimization of a novel structural series of selective HDAC1/HDAC2 inhibitors by the MRL-Boston Parallel Medicinal Chemistry group. An initial lead from a small parallel library was found to be potent and selective in biochemical assays. Advanced compounds were the culmination of iterative library design and possess excellent biochemical and cellular potency, as well as acceptable PK and efficacy in animal models.


Bioorganic & Medicinal Chemistry Letters | 2009

Exploring the pharmacokinetic properties of phosphorus-containing selective HDAC 1 and 2 inhibitors (SHI-1:2).

Richard Heidebrecht; Melissa Chenard; Joshua Close; William K. Dahlberg; Judith C. Fleming; Jonathan Grimm; Julie E. Hamill; Andreas Harsch; Brian B. Haines; Bethany Hughes; Astrid M. Kral; Richard E. Middleton; Chandrasekhar Mushti; Nicole Ozerova; Alexander A. Szewczak; Hongmei Wang; Kevin J. Wilson; David J. Witter; J. Paul Secrist; Thomas A. Miller

We report the preparation and structure-activity relationships of phosphorus-containing histone deacetylase inhibitors. A strong trend between decreasing phosphorus functional group size and superior mouse pharmacokinetic properties was identified. In addition, optimized candidates showed tumor growth inhibition in xenograft studies.


Journal of Pharmacological and Toxicological Methods | 2011

Direct determination of the ratio of unbound fraction in plasma to unbound fraction in microsomal system (fup/fumic) for refined prediction of phase I mediated metabolic hepatic clearance

Sujal V. Deshmukh; Andreas Harsch

At the drug discovery stage, in vivo metabolic hepatic clearance (CL(hep)) is commonly predicted using in vitro parent compound disappearance data generated in liver microsomes or hepatocytes. Correction for the unbound fraction of a compound in the in vitro system and in plasma/serum is known to be critical for the accuracy of metabolic clearance predictions. Discrete generation of these required experimental parameters can be laborious. Herein, we describe a straightforward and direct approach to obtain the ratio of unbound fraction in plasma (fu(p)) to unbound fraction in the microsomal system (fu(mic)) of a small molecule compound using equilibrium dialysis. Experimental conditions were optimized with respect to incubation time, temperature, and plate shaking speed. Results obtained from this system were validated for a set of test compounds by comparison to individually measured fu(p) and fu(mic) data using ultracentrifugation. The correlation for fu(p)/fu(mic) between the two methods for a set of 23 data points was very good with R(2) of 0.94, slope of 1.05 and an intercept of 0.007. The impact of microsomal binding on predicted CL(hep) was illustrated for a tightly bound compound using a series of incubations with increasing concentration of monkey liver microsomal protein. Alteration of this experimental parameter profoundly affected calculated CL(hep) using the well-stirred model. Significant differences were observed in the prediction when the model was corrected for fu(p) only; in contrast, the model corrected for plasma protein and microsomal protein binding predicted clearance values independent of the microsomal protein concentration.


ACS Medicinal Chemistry Letters | 2014

Delayed and Prolonged Histone Hyperacetylation with a Selective HDAC1/HDAC2 Inhibitor.

Joey L. Methot; Dawn Mampreian Hoffman; David J. Witter; Matthew G. Stanton; Paul Harrington; Christopher Hamblett; Phieng Siliphaivanh; Kevin J. Wilson; Jed L. Hubbs; Richard Heidebrecht; Astrid M. Kral; Nicole Ozerova; Judith C. Fleming; Hongmei Wang; Alexander A. Szewczak; Richard E. Middleton; Bethany Hughes; Jonathan C. Cruz; Brian B. Haines; Melissa Chenard; Candia M. Kenific; Andreas Harsch; J. Paul Secrist; Thomas A. Miller

The identification and in vitro and in vivo characterization of a potent SHI-1:2 are described. Kinetic analysis indicated that biaryl inhibitors exhibit slow binding kinetics in isolated HDAC1 and HDAC2 preparations. Delayed histone hyperacetylation and gene expression changes were also observed in cell culture, and histone acetylation was observed in vivo beyond disappearance of drug from plasma. In vivo studies further demonstrated that continuous target inhibition was well tolerated and efficacious in tumor-bearing mice, leading to tumor growth inhibition with either once-daily or intermittent administration.


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.


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.

Researchain Logo
Decentralizing Knowledge