Network


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

Hotspot


Dive into the research topics where Michael R. Hale is active.

Publication


Featured researches published by Michael R. Hale.


Nature Chemical Biology | 2015

Translating slow-binding inhibition kinetics into cellular and in vivo effects

Grant K. Walkup; Zhiping You; Philip L. Ross; Eleanor K. H. Allen; Fereidoon Daryaee; Michael R. Hale; John P. O'Donnell; David E. Ehmann; Virna J. A. Schuck; Ed T. Buurman; Allison L Choy; Laurel Hajec; Kerry E. Murphy-Benenato; Valerie Marone; Sara A. Patey; Lena A Grosser; Stephen G. Walker; Peter J. Tonge; Stewart L. Fisher

Many drug candidates fail in clinical trials owing to a lack of efficacy from limited target engagement or an insufficient therapeutic index. Minimizing off-target effects while retaining the desired pharmacodynamic (PD) response can be achieved by reduced exposure for drugs that display kinetic selectivity in which the drug-target complex has a longer half-life than off-target-drug complexes. However, though slow-binding inhibition kinetics are a key feature of many marketed drugs, prospective tools that integrate drug-target residence time into predictions of drug efficacy are lacking, hindering the integration of drug-target kinetics into the drug discovery cascade. Here we describe a mechanistic PD model that includes drug-target kinetic parameters, including the on- and off-rates for the formation and breakdown of the drug-target complex. We demonstrate the utility of this model by using it to predict dose response curves for inhibitors of the LpxC enzyme from Pseudomonas aeruginosa in an animal model of infection.


Toxicological Sciences | 2014

Early Prediction of Polymyxin-Induced Nephrotoxicity With Next-Generation Urinary Kidney Injury Biomarkers

Natalie Keirstead; Matthew Wagoner; Patricia Bentley; Marie Blais; Crystal Brown; Letitia Cheatham; Paul Ciaccio; Yvonne P. Dragan; Douglas Ferguson; Jim Fikes; Melanie Galvin; Anshul Gupta; Michael R. Hale; Nakpangi Johnson; Wenli Luo; Frank McGrath; Mark Pietras; Sally A. Price; Abhishek Sathe; Jennifer C. Sasaki; Debra Snow; Robert L. Walsky; Gunther Kern

Despite six decades of clinical experience with the polymyxin class of antibiotics, their dose-limiting nephrotoxicity remains difficult to predict due to a paucity of sensitive biomarkers. Here, we evaluate the performance of standard of care and next-generation biomarkers of renal injury in the detection and monitoring of polymyxin-induced acute kidney injury in male Han Wistar rats using colistin (polymyxin E) and a polymyxin B (PMB) derivative with reduced nephrotoxicity, PMB nonapeptide (PMBN). This study provides the first histopathological and biomarker analysis of PMBN, an important test of the hypothesis that fatty acid modifications and charge reductions in polymyxins can reduce their nephrotoxicity. The results indicate that alterations in a panel of urinary kidney injury biomarkers can be used to monitor histopathological injury, with Kim-1 and α-GST emerging as the most sensitive biomarkers outperforming clinical standards of care, serum or plasma creatinine and blood urea nitrogen. To enable the prediction of polymyxin-induced nephrotoxicity, an in vitro cytotoxicity assay was employed using human proximal tubule epithelial cells (HK-2). Cytotoxicity data in these HK-2 cells correlated with the renal toxicity detected via safety biomarker data and histopathological evaluation, suggesting that in vitro and in vivo methods can be incorporated within a screening cascade to prioritize polymyxin class analogs with more favorable renal toxicity profiles.


Bioorganic & Medicinal Chemistry Letters | 2013

Exploring the UDP pocket of LpxC through amino acid analogs.

Michael R. Hale; Pamela Hill; Sushmita D. Lahiri; Matthew D. Miller; Philip L. Ross; Richard A. Alm; Ning Gao; Amy Kutschke; Bryan Prince; Jason Thresher; Wei Yang

Lipopolysaccharide (LPS) biosynthesis is an attractive antibacterial target as it is both conserved and essential for the survival of key pathogenic bacteria. Lipid A is the hydrophobic anchor for LPS and a key structural component of the outer membrane of Gram-negative bacteria. Lipid A biosynthesis is performed in part by a unique zinc dependent metalloamidase, LpxC (UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase), which catalyzes the first non-reversible step in lipid A biosynthesis. The UDP portion of the LpxC substrate-binding pocket has been relatively unexplored. We have designed and evaluated a series of hydroxamate based inhibitors which explore the SAR of substitutions directed into the UDP pocket with a range of substituted α-amino acid based linkers. We also provide the first wild type structure of Pseudomonas aeruginosa LpxC which was utilized in the design of many of these analogs.


ACS Medicinal Chemistry Letters | 2014

Synthesis, Structure, and SAR of Tetrahydropyran-Based LpxC Inhibitors

Kerry E. Murphy-Benenato; Nelson B. Olivier; Allison L Choy; Philip L. Ross; Matthew D. Miller; Jason Thresher; Ning Gao; Michael R. Hale

In the search for novel Gram-negative agents, we performed a comprehensive search of the AstraZeneca collection and identified a tetrahydropyran-based matrix metalloprotease (MMP) inhibitor that demonstrated nanomolar inhibition of UDP-3-O-(acyl)-N-acetylglucosamine deacetylase (LpxC). Crystallographic studies in Aquifex aeolicus LpxC indicated the tetrahydropyran engaged in the same hydrogen bonds and van der Waals interactions as other known inhibitors. Systematic optimization of three locales on the scaffold provided compounds with improved Gram-negative activity. However, the optimization of LpxC activity was not accompanied by reduced inhibition of MMPs. Comparison of the crystal structure of the native product, UDP-3-O-(acyl)-glucosamine, in Aquifex aeolicus to the structure of a tetrahydropyran-based inhibitor indicates pathways for future optimization.


Cancer Research | 2015

Abstract 4693: The selective ERK inhibitor BVD-523 is active in models of MAPK pathway-dependent cancers, including those with intrinsic and acquired drug resistance

Ursula A. Germann; Brinley Furey; Jeff Roix; William Markland; Russell Hoover; Alex Aronov; Michael R. Hale; Guanjing Chen; Gabriel Martinez-Botella; Rossitza Gueorguieva Alargova; Bin Fan; David Sorrell; Kay R. Meshaw; Paul Shapiro; Michael J. Wick; Cyril H. Benes; Mathew J. Garnett; Gary A Decrescenzo; Mark Namchuk; Saurabh Saha; Dean Welsch

The MAPK (RAS-RAF-MEK-ERK) pathway is activated in many cancers, and the clinical efficacy of BRAF and MEK inhibitors in melanoma confirms that targeting the MAPK pathway has therapeutic potential. Unfortunately, intrinsic and acquired drug resistance limits use of MAPK-directed therapies, and resistance is often associated with activated ERK signaling. Here, we report characterization of BVD-523 (ulixertinib), a novel small-molecule ERK1/2 kinase inhibitor currently under investigation in Phase 1 clinical trials. BVD-523 potently and selectively inhibits ERK1 and ERK2 kinases in a reversible, ATP-competitive fashion. Consistent with its mechanism of action, BVD-523 inhibits signal transduction, cell proliferation, and cell survival, most potently in cell lines bearing mutations that activate MAPK pathway signaling. Similarly, single-agent BVD-523 inhibits tumor growth in vivo in BRAF-mutant melanoma and colorectal xenografts as well as in KRAS-mutant colorectal and pancreatic models. Combination treatment with BVD-523 and dabrafenib inhibits tumor growth in a BRAF-mutant melanoma model. Importantly, BVD-523 is effective in several models that show intrinsic or acquired resistance to other MAPK pathway inhibitors. BVD-523 inhibits with equivalent potency the growth of parental cells or those cultured for resistance to dabrafenib, trametinib, or the combination of both drugs. Additionally, BVD-523 inhibits growth in wild-type cells and a RAF/MEK cross-resistant cell line bearing a MEK1 Q56P mutation with similar potency. Lastly, single-agent BVD-523 inhibits the growth of a patient-derived tumor xenograft harboring cross-resistance to dabrafenib, trametinib, and the combination treatment following clinical progression on a MEK inhibitor. Phase 1 trials of BVD-523 are currently recruiting patients with advanced solid tumors (NCT0178429) or hematologic malignancies (NCT02296242). Eligibility criteria include diagnosis according to certain genetic features, and treatment in backgrounds including progression following prior MAPK targeted therapy. The primary objective of these studies is to identify the recommended Phase 2 dose(s) for single-agent BVD-523 treatment. Additional objectives include pharmacokinetic and pharmacodynamic assessments, and preliminary measures of efficacy. The solid tumor protocol has met its study objectives in Part 1 (defining the safety profile and maximum tolerated dose), and will be reported separately; findings appear consistent with the activity profile defined in preclinical studies. In total, preclinical and clinical studies will help elucidate how BVD-523 (ulixertinib) may be used as a novel agent in MAPK-directed therapeutic strategies, including for patients that have failed treatment due to intrinsic or acquired resistance and active signaling through ERK. Citation Format: Ursula Germann, Brinley Furey, Jeff Roix, William Markland, Russell Hoover, Alex Aronov, Michael Hale, Guanjing Chen, Gabriel Martinez-Botella, Rossitza Alargova, Bin Fan, David Sorrell, Kay Meshaw, Paul Shapiro, Michael J. Wick, Cyril Benes, Mathew Garnett, Gary DeCrescenzo, Mark Namchuk, Saurabh Saha, Dean J. Welsch. The selective ERK inhibitor BVD-523 is active in models of MAPK pathway-dependent cancers, including those with intrinsic and acquired drug resistance. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4693. doi:10.1158/1538-7445.AM2015-4693


Bioorganic & Medicinal Chemistry Letters | 2015

Negishi cross-coupling enabled synthesis of novel NAD(+)-dependent DNA ligase inhibitors and SAR development.

Kerry E. Murphy-Benenato; Lakshmaiah Gingipalli; P. Ann Boriack-Sjodin; Gabriel Martinez-Botella; Dan Carcanague; Charles J. Eyermann; Madhu Gowravaram; Jenna Harang; Michael R. Hale; Georgine Ioannidis; Harris Jahic; Amy Kutschke; Valerie A. Laganas; James T. Loch; Matthew D. Miller; Herbert Oguto; Sahil Joe Patel

Two novel compounds, pyridopyrimidines (1) and naphthyridines (2) were identified as potent inhibitors of bacterial NAD(+)-dependent DNA ligase (Lig) A in a fragment screening. SAR was guided by molecular modeling and X-ray crystallography. It was observed that the diaminonitrile pharmacophore made a key interaction with the ligase enzyme, specifically residues Glu114, Lys291, and Leu117. Synthetic challenges limited opportunities for diversification of the naphthyridine core, therefore most of the SAR was focused on a pyridopyrimidine scaffold. The initial diversification at R(1) improved both enzyme and cell potency. Further SAR developed at the R(2) position using the Negishi cross-coupling reaction provided several compounds, among these compounds 22g showed good enzyme potency and cellular potency.


Protein Expression and Purification | 2014

Overexpression of Pseudomonas aeruginosa LpxC with its inhibitors in an acrB-deficient Escherichia coli strain

Ning Gao; Sarah M. McLeod; Laurel Hajec; Nelson B. Olivier; Sushmita D. Lahiri; D. Bryan Prince; Jason Thresher; Philip L. Ross; James Whiteaker; Peter Doig; Amanda Haixi Li; Pamela Hill; Mark Cornebise; Folkert Reck; Michael R. Hale

In Gram-negative bacteria, the cell wall is surrounded by an outer membrane, the outer leaflet of which is comprised of charged lipopolysaccharide (LPS) molecules. Lipid A, a component of LPS, anchors this molecule to the outer membrane. UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase (LpxC) is a zinc-dependent metalloamidase that catalyzes the first committed step of biosynthesis of Lipid A, making it a promising target for antibiotic therapy. Formation of soluble aggregates of Pseudomonas aeruginosa LpxC protein when overexpressed in Escherichia coli has limited the availability of high quality protein for X-ray crystallography. Expression of LpxC in the presence of an inhibitor dramatically increased protein solubility, shortened crystallization time and led to a high-resolution crystal structure of LpxC bound to the inhibitor. However, this approach required large amounts of compound, restricting its use. To reduce the amount of compound needed, an overexpression strain of E. coli was created lacking acrB, a critical component of the major efflux pump. By overexpressing LpxC in the efflux deficient strain in the presence of LpxC inhibitors, several structures of P. aeruginosa LpxC in complex with different compounds were solved to accelerate structure-based drug design.


Archive | 2010

Hydroxamic acid derivatives as gram-negative antibacterial agents

Kerry Ellen Benenato; Allison Laura Choy; Michael R. Hale; Pamela Hill; Valerie Marone; Matthew D. Miller


Tetrahedron Letters | 2015

From Fragments to Leads: Novel Bacterial Nad+-Dependent DNA Ligase Inhibitors

Michael R. Hale; Claire Brassington; Dan Carcanague; Kevin J. Embrey; Charles J. Eyermann; Robert A. Giacobbe; Lakshmaiah Gingipalli; Madhusudhan Gowravaram; Jenna Harang; Tina Howard; Georgine Ioannidis; Haris Jahić; Amy Kutschke; Valerie A. Laganas; James T. Loch; Matthew D. Miller; Kerry E. Murphy-Benenato; Herbert Oguto; Ludovic R. Otterbein; Sahil Joe Patel; Adam B. Shapiro; P. Ann Boriack-Sjodin


The FASEB Journal | 2015

Toxicokinetic Insights are Critical to Understanding Renal Toxicity of Novel Polymyxin Analog

Anshul Gupta; Natalie Keirstead; Harish Shankaran; Richard J. A. Goodwin; Aixiang Xue; Kim Martea; Nakpangi Johnson; Tyler Grebe; Michael R. Hale; Gunther Kern; John G. Swales; Jennifer C. Sasaki

Collaboration


Dive into the Michael R. Hale's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge