Network


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

Hotspot


Dive into the research topics where Astrid M. Kral is active.

Publication


Featured researches published by Astrid M. Kral.


Nature Medicine | 1995

Inhibition of farnesyltransferase induces regression of mammary and salivary carcinomas in ras transgenic mice.

Nancy E. Kohl; Charles A. Omer; Michael W. Conner; Neville J. Anthony; Joseph P. Davide; S. Jane Desolms; Elizabeth A. Giuliani; Robert P. Gomez; Samuel L. Graham; Kelly Hamilton; Laurence K. Handt; George D. Hartman; Kenneth S. Koblan; Astrid M. Kral; Patricia Miller; Scott D. Mosser; Timothy J. O'Neill; Elaine Rands; Michael D. Schaber; Jackson B. Gibbs; Allen Oliff

For Ras oncoproteins to transform mammalian cells, they must be post-translationally modified with a farnesyl group in a reaction catalysed by the enzyme farnesyl-protein transferase (FPTase). Inhibitors of FPTase have therefore been proposed as anti-cancer agents. We show that L-744,832, which mimics the CaaX motif to which the farnesyl group is added, is a potent and selective inhibitor of FPTase. In MMTV-v-Ha-ras mice bearing palpable tumours, daily administration of L-744,832 caused tumour regression. Following cessation of treatment, tumours reappeared, the majority of which regressed upon retreatment. No systemic toxicity was found upon necropsy of L-744,832-treated mice. This first demonstration of anti-FPTase-mediated tumour regression suggests that FPTase inhibitors may be safe and effective anti-tumour agents in some cancers.


Biochemical Journal | 2005

Identification and characterization of pleckstrin-homology-domain-dependent and isoenzyme-specific Akt inhibitors

Stanley F. Barnett; Deborah Defeo-Jones; Sheng Fu; Paula J. Hancock; Kathleen M. Haskell; Raymond E. Jones; Jason Kahana; Astrid M. Kral; Karen R. Leander; Ling L. Lee; John Malinowski; Elizabeth McAvoy; Debbie D. Nahas; Ronald G. Robinson; Hans E. Huber

We developed a high-throughput HTRF (homogeneous time-resolved fluorescence) assay for Akt kinase activity and screened approx. 270000 compounds for their ability to inhibit the three isoforms of Akt. Two Akt inhibitors were identified that exhibited isoenzyme specificity. The first compound (Akt-I-1) inhibited only Akt1 (IC50 4.6 microM) while the second compound (Akt-I-1,2) inhibited both Akt1 and Akt2 with IC50 values of 2.7 and 21 microM respectively. Neither compound inhibited Akt3 nor mutants lacking the PH (pleckstrin homology) domain at concentrations up to 250 microM. These compounds were reversible inhibitors, and exhibited a linear mixed-type inhibition against ATP and peptide substrate. In addition to inhibiting kinase activity of individual Akt isoforms, both inhibitors blocked the phosphorylation and activation of the corresponding Akt isoforms by PDK1 (phosphoinositide-dependent kinase 1). A model is proposed in which these inhibitors bind to a site formed only in the presence of the PH domain. Binding of the inhibitor is postulated to promote the formation of an inactive conformation. In support of this model, antibodies to the Akt PH domain or hinge region blocked the inhibition of Akt by Akt-I-1 and Akt-I-1,2. These inhibitors were found to be cell-active and to block phosphorylation of Akt at Thr308 and Ser473, reduce the levels of active Akt in cells, block the phosphorylation of known Akt substrates and promote TRAIL (tumour-necrosis-factor-related apoptosis-inducing ligand)-induced apoptosis in LNCap prostate cancer cells.


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.


Proceedings of the National Academy of Sciences of the United States of America | 2001

The crystal structure of human protein farnesyltransferase reveals the basis for inhibition by CaaX tetrapeptides and their mimetics

Stephen B. Long; Paula J. Hancock; Astrid M. Kral; Homme W. Hellinga; Lorena S. Beese

Protein farnesyltransferase (FTase) catalyzes the attachment of a farnesyl lipid group to the cysteine residue located in the C-terminal tetrapeptide of many essential signal transduction proteins, including members of the Ras superfamily. Farnesylation is essential both for normal functioning of these proteins, and for the transforming activity of oncogenic mutants. Consequently FTase is an important target for anti-cancer therapeutics. Several FTase inhibitors are currently undergoing clinical trials for cancer treatment. Here, we present the crystal structure of human FTase, as well as ternary complexes with the TKCVFM hexapeptide substrate, CVFM non-substrate tetrapeptide, and L-739,750 peptidomimetic with either farnesyl diphosphate (FPP), or a nonreactive analogue. These structures reveal the structural mechanism of FTase inhibition. Some CaaX tetrapeptide inhibitors are not farnesylated, and are more effective inhibitors than farnesylated CaaX tetrapeptides. CVFM and L-739,750 are not farnesylated, because these inhibitors bind in a conformation that is distinct from the TKCVFM hexapeptide substrate. This non-substrate binding mode is stabilized by an ion pair between the peptide N terminus and the α-phosphate of the FPP substrate. Conformational mapping calculations reveal the basis for the sequence specificity in the third position of the CaaX motif that determines whether a tetrapeptide is a substrate or non-substrate. The presence of β-branched amino acids in this position prevents formation of the non-substrate conformation; all other aliphatic amino acids in this position are predicted to form the non-substrate conformation, provided their N terminus is available to bind to the FPP α-phosphate. These results may facilitate further development of FTase inhibitors.


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

Rapid assembly of diverse and potent allosteric Akt inhibitors.

Zhicai Wu; Ronald G. Robinson; Sheng Fu; Stanley F. Barnett; Deborah Defeo-Jones; Raymond E. Jones; Astrid M. Kral; Hans E. Huber; Nancy E. Kohl; George D. Hartman; Mark T. Bilodeau

This paper describes the rapid assembly of four different classes of potent Akt inhibitors from a common intermediate. Among them, a pyridopyrimidine series displayed the best intrinsic and cell potency against Akt1 and Akt2. This series also showed a promising pharmacokinetic profile and excellent selectivity over other closely related kinases.


Molecular Cancer Therapeutics | 2013

Molecular and biologic analysis of histone deacetylase inhibitors with diverse specificities.

Andrea Newbold; Geoffrey M. Matthews; Michael Bots; Leonie A. Cluse; Christopher J. Clarke; Kellie M. Banks; Carleen Cullinane; Jessica E. Bolden; Ailsa J. Christiansen; Ross A. Dickins; Claudia Miccolo; Susanna Chiocca; Astrid M. Kral; Nicole Ozerova; Thomas A. Miller; Joey L. Methot; Victoria M. Richon; J. Paul Secrist; Saverio Minucci; Ricky W. Johnstone

Histone deacetylase inhibitors (HDACi) are anticancer agents that induce hyperacetylation of histones, resulting in chromatin remodeling and transcriptional changes. In addition, nonhistone proteins, such as the chaperone protein Hsp90, are functionally regulated through hyperacetylation mediated by HDACis. Histone acetylation is thought to be primarily regulated by HDACs 1, 2, and 3, whereas the acetylation of Hsp90 has been proposed to be specifically regulated through HDAC6. We compared the molecular and biologic effects induced by an HDACi with broad HDAC specificity (vorinostat) with agents that predominantly inhibited selected class I HDACs (MRLB-223 and romidepsin). MRLB-223, a potent inhibitor of HDACs 1 and 2, killed tumor cells using the same apoptotic pathways as the HDAC 1, 2, 3, 6, and 8 inhibitor vorinostat. However, vorinostat induced histone hyperacetylation and killed tumor cells more rapidly than MRLB-223 and had greater therapeutic efficacy in vivo. FDCP-1 cells dependent on the Hsp90 client protein Bcr-Abl for survival, were killed by all HDACis tested, concomitant with caspase-dependent degradation of Bcr-Abl. These studies provide evidence that inhibition of HDAC6 and degradation of Bcr-Abl following hyperacetylation of Hsp90 is likely not a major mechanism of action of HDACis as had been previously posited. Mol Cancer Ther; 12(12); 2709–21. ©2013 AACR.


Bioorganic & Medicinal Chemistry Letters | 2008

Optimization of 2,3,5-trisubstituted pyridine derivatives as potent allosteric Akt1 and Akt2 inhibitors

John C. Hartnett; Stanley F. Barnett; Mark T. Bilodeau; Deborah Defeo-Jones; George D. Hartman; Hans E. Huber; Raymond E. Jones; Astrid M. Kral; Ronald G. Robinson; Zhicai Wu

This letter shows inhibitor SAR on a pyridine series of allosteric Akt inhibitors to optimize enzymatic and cellular potency. We have optimized 2,3,5-trisubstituted pyridines to give potent Akt1 and Akt2 inhibitors in both enzyme and cell based assays. In addition, we will also highlight the pharmacokinetic profile of an optimized inhibitor that has low clearance and long half-life in dogs.


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.

Researchain Logo
Decentralizing Knowledge