Network


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

Hotspot


Dive into the research topics where Michael A. Stashko is active.

Publication


Featured researches published by Michael A. Stashko.


Journal of Medicinal Chemistry | 2014

UNC2025, a Potent and Orally Bioavailable MER/FLT3 Dual Inhibitor

Weihe Zhang; Deborah DeRyckere; Debra Hunter; Jing Liu; Michael A. Stashko; Katherine A. Minson; Christopher T. Cummings; Trevor G. Glaros; Dianne Newton; Susan Sather; Dehui Zhang; Dmitri Kireev; William P. Janzen; H. Shelton Earp; Douglas K. Graham; Stephen V. Frye; Xiaodong Wang

We previously reported a potent small molecule Mer tyrosine kinase inhibitor UNC1062. However, its poor PK properties prevented further assessment in vivo. We report here the sequential modification of UNC1062 to address DMPK properties and yield a new potent and highly orally bioavailable Mer inhibitor, 11, capable of inhibiting Mer phosphorylation in vivo, following oral dosing as demonstrated by pharmaco-dynamic (PD) studies examining phospho-Mer in leukemic blasts from mouse bone marrow. Kinome profiling versus more than 300 kinases in vitro and cellular selectivity assessments demonstrate that 11 has similar subnanomolar activity against Flt3, an additional important target in acute myelogenous leukemia (AML), with pharmacologically useful selectivity versus other kinases examined.


European Journal of Medicinal Chemistry | 2013

UNC1062, a new and potent Mer inhibitor.

Jing Liu; Weihe Zhang; Michael A. Stashko; Deborah DeRyckere; Christopher T. Cummings; Debra Hunter; Chao Yang; Chatura N. Jayakody; Nancy Cheng; Catherine Simpson; Jacqueline Norris-Drouin; Susan Sather; Dmitri Kireev; William P. Janzen; H. Shelton Earp; Douglas K. Graham; Stephen V. Frye; Xiaodong Wang

Abnormal activation of Mer kinase has been implicated in the oncogenesis of many human cancers including acute lymphoblastic and myeloid leukemia, non-small cell lung cancer, and glioblastoma. We have discovered a new family of small molecule Mer inhibitors, pyrazolopyrimidine sulfonamides, that potently inhibit the kinase activity of Mer. Importantly, these compounds do not demonstrate significant hERG activity in the PatchXpress assay. Through structure-activity relationship studies, 35 (UNC1062) was identified as a potent (IC50 = 1.1 nM) and selective Mer inhibitor. When applied to live tumor cells, UNC1062 inhibited Mer phosphorylation and colony formation in soft agar. Given the potential of Mer as a therapeutic target, UNC1062 is a promising candidate for further drug development.


Journal of Medicinal Chemistry | 2013

Discovery of Mer Specific Tyrosine Kinase Inhibitors for the Treatment and Prevention of Thrombosis

Weihe Zhang; Andrew L. McIver; Michael A. Stashko; Deborah DeRyckere; Brian R. Branchford; Debra Hunter; Dmitri Kireev; Michael J. Miley; Jacqueline Norris-Drouin; Wendy M. Stewart; Susan Sather; Yingqiu Zhou; Jorge Di Paola; Mischa Machius; William P. Janzen; H. Shelton Earp; Douglas K. Graham; Stephen V. Frye; Xiaodong Wang

The role of Mer kinase in regulating the second phase of platelet activation generates an opportunity to use Mer inhibitors for preventing thrombosis with diminished likelihood for bleeding as compared to current therapies. Toward this end, we have discovered a novel, Mer kinase specific substituted-pyrimidine scaffold using a structure-based drug design and a pseudo ring replacement strategy. The cocrystal structure of Mer with two compounds (7 and 22) possessing distinct activity have been determined. Subsequent SAR studies identified compound 23 (UNC2881) as a lead compound for in vivo evaluation. When applied to live cells, 23 inhibits steady-state Mer kinase phosphorylation with an IC50 value of 22 nM. Treatment with 23 is also sufficient to block EGF-mediated stimulation of a chimeric receptor containing the intracellular domain of Mer fused to the extracellular domain of EGFR. In addition, 23 potently inhibits collagen-induced platelet aggregation, suggesting that this class of inhibitors may have utility for prevention and/or treatment of pathologic thrombosis.


Journal of Medicinal Chemistry | 2013

Pseudo-Cyclization through Intramolecular Hydrogen Bond Enables Discovery of Pyridine Substituted Pyrimidines as New Mer Kinase Inhibitors

Weihe Zhang; Dehui Zhang; Michael A. Stashko; Deborah DeRyckere; Debra Hunter; Dmitri Kireev; Michael J. Miley; Christopher T. Cummings; Jacqueline Norris-Drouin; Wendy M. Stewart; Susan Sather; Yingqiu Zhou; Gregory Kirkpatrick; Mischa Machius; William P. Janzen; H. Shelton Earp; Douglas K. Graham; Stephen V. Frye; Xiaodong Wang

Abnormal activation or overexpression of Mer receptor tyrosine kinase has been implicated in survival signaling and chemoresistance in many human cancers. Consequently, Mer is a promising novel cancer therapeutic target. A structure-based drug design approach using a pseudo-ring replacement strategy was developed and validated to discover a new family of pyridinepyrimidine analogues as potent Mer inhibitors. Through SAR studies, 10 (UNC2250) was identified as the lead compound for further investigation based on high selectivity against other kinases and good pharmacokinetic properties. When applied to live cells, 10 inhibited steady-state phosphorylation of endogenous Mer with an IC50 of 9.8 nM and blocked ligand-stimulated activation of a chimeric EGFR-Mer protein. Treatment with 10 also resulted in decreased colony-forming potential in rhabdoid and NSCLC tumor cells, thereby demonstrating functional antitumor activity. The results provide a rationale for further investigation of this compound for therapeutic application in patients with cancer.


Neuron | 2014

The lipid kinase PIP5K1C regulates pain signaling and sensitization.

Brittany D. Wright; Lipin Loo; Sarah E. Street; Anqi Ma; Bonnie Taylor-Blake; Michael A. Stashko; Jian Jin; William P. Janzen; Stephen V. Frye; Mark J. Zylka

Numerous pain-producing (pronociceptive) receptors signal via phosphatidylinositol 4,5-bisphosphate (PIP2) hydrolysis. However, it is currently unknown which lipid kinases generate PIP2 in nociceptive dorsal root ganglia (DRG) neurons and if these kinases regulate pronociceptive receptor signaling. Here, we found that phosphatidylinositol 4-phosphate 5 kinase type 1C (PIP5K1C) is expressed at higher levels than any other PIP5K and, based on experiments with Pip5k1c(+/-) mice, generates at least half of all PIP2 in DRG neurons. Additionally, Pip5k1c haploinsufficiency reduces pronociceptive receptor signaling and TRPV1 sensitization in DRG neurons as well as thermal and mechanical hypersensitivity in mouse models of chronic pain. We identified a small molecule inhibitor of PIP5K1C (UNC3230) in a high-throughput screen. UNC3230 lowered PIP2 levels in DRG neurons and attenuated hypersensitivity when administered intrathecally or into the hindpaw. Our studies reveal that PIP5K1C regulates PIP2-dependent nociceptive signaling and suggest that PIP5K1C is a therapeutic target for chronic pain.


JCI insight | 2016

The MERTK/FLT3 inhibitor MRX-2843 overcomes resistance-conferring FLT3 mutations in acute myeloid leukemia

Katherine A. Minson; Catherine C. Smith; Deborah DeRyckere; Clara Libbrecht; Alisa B. Lee-Sherick; Madeline G. Huey; Elisabeth Lasater; Gregory Kirkpatrick; Michael A. Stashko; Weihe Zhang; Craig T. Jordan; Dmitri Kireev; Xiaodong Wang; Stephen V. Frye; H. Shelton Earp; Neil P. Shah; Douglas K. Graham

FMS-like tyrosine kinase 3-targeted (FLT3-targeted) therapies have shown initial promise for the treatment of acute myeloid leukemia (AML) expressing FLT3-activating mutations; however, resistance emerges rapidly. Furthermore, limited options exist for the treatment of FLT3-independent AML, demonstrating the need for novel therapies that reduce toxicity and improve survival. MERTK receptor tyrosine kinase is overexpressed in 80% to 90% of AMLs and contributes to leukemogenesis. Here, we describe MRX-2843, a type 1 small-molecule tyrosine kinase inhibitor that abrogates activation of both MERTK and FLT3 and their downstream effectors. MRX-2843 treatment induces apoptosis and inhibits colony formation in AML cell lines and primary patient samples expressing MERTK and/or FLT3-ITD, with a wide therapeutic window compared with that of normal human cord blood cells. In murine orthotopic xenograft models, once-daily oral therapy prolonged survival 2- to 3-fold over that of vehicle-treated controls. Additionally, MRX-2843 retained activity against quizartinib-resistant FLT3-ITD-mutant proteins with clinically relevant alterations at the D835 or F691 loci and prolonged survival in xenograft models of quizartinib-resistant AML. Together, these observations validate MRX-2843 as a translational agent and support its clinical development for the treatment of AML.


Journal of Biomolecular Screening | 2015

Development of a High-Throughput Screening Assay to Identify Inhibitors of the Lipid Kinase PIP5K1C

Brittany D. Wright; Catherine Simpson; Michael A. Stashko; Dmitri Kireev; Emily A. Hull-Ryde; Mark J. Zylka; William P. Janzen

Phosphatidylinositol 4-phosphate 5-kinases (PIP5Ks) regulate a variety of cellular processes, including signaling through G protein-coupled receptors (GPCRs), endocytosis, exocytosis, and cell migration. These lipid kinases synthesize phosphatidylinositol 4,5-bisphosphate (PIP2) from phosphatidylinositol 4-phosphate [PI(4)P]. Because small-molecule inhibitors of these lipid kinases did not exist, molecular and genetic approaches were predominantly used to study PIP5K1 regulation of these cellular processes. Moreover, standard radioisotope-based lipid kinase assays cannot be easily adapted for high-throughput screening. Here, we report a novel, high-throughput, microfluidic mobility shift assay to identify inhibitors of PIP5K1C. This assay uses fluorescently labeled phosphatidylinositol 4-phosphate as the substrate and recombinant human PIP5K1C. Our assay exhibited high reproducibility, had a calculated adenosine triphosphate Michaelis constant (Km) of 15 µM, performed with z’ values >0.7, and was used to screen a kinase-focused library of ~4700 compounds. From this screen, we identified several potent inhibitors of PIP5K1C, including UNC3230, a compound that we recently found can reduce nociceptive sensitization in animal models of chronic pain. This novel assay will allow continued drug discovery efforts for PIP5K1C and can be adapted easily to screen additional lipid kinases.


Bioorganic & Medicinal Chemistry | 2015

Discovery of Mer kinase inhibitors by Virtual Screening using Structural Protein-Ligand Interaction Fingerprints

C. Da; Michael A. Stashko; Chatura N. Jayakody; Xiaodong Wang; William P. Janzen; Stephen V. Frye; Dmitri Kireev

Mer is a receptor tyrosine kinase implicated in acute lymphoblastic leukemia (ALL), the most common malignancy in children. The currently available data provide a rationale for development of Mer kinase inhibitors as cancer therapeutics that can target both cell autologous and immune-modulatory anti-tumor effects. We have previously reported several series of potent Mer inhibitors and the objective of the current report is to identify a chemically dissimilar back-up series that might circumvent potential, but currently unknown, flaws inherent to the lead series. To this end, we virtually screened a database of ∼3.8million commercially available compounds using high-throughput docking followed by a filter involving Structural Protein-Ligand Interaction Fingerprints (SPLIF). SPLIF permits a quantitative assessment of whether a docking pose interacts with the protein target similarly to an endogenous or known synthetic ligand, and therefore helps to improve both sensitivity and specificity with respect to the docking score alone. Of the total of 62 experimentally tested compounds, 15 demonstrated reliable dose-dependent responses in the Mer in vitro kinase activity assay with inhibitory potencies ranging from 0.46μM to 9.9μM.


Bioorganic & Medicinal Chemistry Letters | 1995

AMINO ACID-DERIVED PIPERIDIDES AS NOVEL CCKB LIGANDS WITH ANXIOLYTIC-LIKE PROPERTIES

Mark W. Holladay; Michael J. Bennett; Hao Bai; Jeffrey W. Ralston; James F. Kerwin; Michael A. Stashko; Thomas R. Miller; Alyssa B. O'Neill; Alex M. Nadzan; Jorge D. Brioni; Chun Wel Lin

Abstract The development of a novel series of carbamoylamino acid benzoylpiperidides as CCK B ligands is described. Selected members of the series antagonized CCK 8 -induced calcium mobilization and showed efficacy in the mouse elevated-plus maze, a measure of potential anxiolytic activity.


Bioorganic & Medicinal Chemistry Letters | 1993

Toward developing peptidomimetics: Successful replacement of backbone amide bonds in tetrapeptide-based CCK-A receptor agonists

Kazumi Shiosaki; Chun Wel Lin; M. Robert Leanna; Howard E. Morton; Thomas R. Miller; David G. Witte; Michael A. Stashko; Alex M. Nadzan

Abstract The backbone amide bonds in two series of tetrapeptide-based CCK-A receptor agonists were systematically replaced with the methylene amino isostere. Potent and selective pseudopeptides were identified that will facilitate our understanding of how these peptides interact with the CCK receptor and their structural correlations with other CCK ligands. This information will aid in the eventual development of peptidomimetics.

Collaboration


Dive into the Michael A. Stashko's collaboration.

Top Co-Authors

Avatar

Stephen V. Frye

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Dmitri Kireev

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Xiaodong Wang

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Weihe Zhang

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Douglas K. Graham

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Deborah DeRyckere

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

H. Shelton Earp

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

William P. Janzen

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jacqueline Norris-Drouin

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Susan Sather

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge