Network


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

Hotspot


Dive into the research topics where Betsy S. Pierce is active.

Publication


Featured researches published by Betsy S. Pierce.


Bioorganic & Medicinal Chemistry Letters | 2009

Benzothiophene inhibitors of MK2. Part 1: structure-activity relationships, assessments of selectivity and cellular potency

David R. Anderson; Marvin Jay Meyers; Ravi G. Kurumbail; Nicole Caspers; Gennadiy I. Poda; Scott A. Long; Betsy S. Pierce; Matthew W. Mahoney; Robert J. Mourey

Identification of potent benzothiophene inhibitors of mitogen activated protein kinase-activated protein kinase 2 (MK2), structure-activity relationship (SAR) studies, selectivity assessments against CDK2, cellular potency and mechanism of action are presented. Crystallographic data provide a rationale for the observed MK2 potency as well as selectivity over CDK2 for this class of inhibitors.


Bioorganic & Medicinal Chemistry Letters | 2009

Benzothiophene inhibitors of MK2. Part 2: improvements in kinase selectivity and cell potency.

David R. Anderson; Marvin Jay Meyers; Ravi G. Kurumbail; Nicole Caspers; Gennadiy I. Poda; Scott A. Long; Betsy S. Pierce; Matthew W. Mahoney; Robert J. Mourey; Mihir D. Parikh

Optimization of kinase selectivity for a set of benzothiophene MK2 inhibitors provided analogs with potencies of less than 500 nM in a cell based assay. The selectivity of the inhibitors can be rationalized by examination of X-ray crystal structures of inhibitors bound to MK2.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery of 2-chloro-N-((4,4-difluoro-1-hydroxycyclohexyl)methyl)-5-(5-fluoropyrimidin-2-yl)benzamide as a potent and CNS penetrable P2X7 receptor antagonist

Xiangyang Chen; Betsy S. Pierce; Win Naing; Margaret L. Grapperhaus; Dennis Paul Phillion

Focused SAR studies were carried out around 5-heteroaryl and 1-amide portions of the 2-chlorobenzamide scaffold, resulting in the discovery of a potent, metabolically stable and centrally penetrable antagonist against P2X(7) receptor.


Organic Letters | 2015

Visible-Light-Driven Photocatalytic Initiation of Radical Thiol–Ene Reactions Using Bismuth Oxide

Olugbeminiyi O. Fadeyi; James J. Mousseau; Yiqing Feng; Christophe Allais; Philippe Nuhant; Ming Z. Chen; Betsy S. Pierce; Ralph P. Robinson

A nontoxic and inexpensive photocatalytic initiation of anti-Markovnikov hydrothiolation of olefins using visible light is reported. This method is characterized by low catalyst loading, thereby enabling a mild and selective method for radical initiation in thiol-ene reactions between a wide scope of olefins and thiols.


Journal of Medicinal Chemistry | 2012

Maximizing lipophilic efficiency: the use of Free-Wilson analysis in the design of inhibitors of acetyl-CoA carboxylase.

Kevin Daniel Freeman-Cook; Paul Amor; Scott Bader; Leanne M. Buzon; Steven B. Coffey; Jeffrey W. Corbett; Kenneth J. DiRico; Shawn D. Doran; Richard L. Elliott; William Esler; Angel Guzman-Perez; Kevin E. Henegar; Janet A. Houser; Christopher S. Jones; Chris Limberakis; Katherine Loomis; Kirk McPherson; Sharad Murdande; Kendra Louise Nelson; Dennis Paul Phillion; Betsy S. Pierce; Wei Song; Eliot Sugarman; Susan Tapley; Meihua Tu; Zhengrong Zhao

This paper describes the design and synthesis of a novel series of dual inhibitors of acetyl-CoA carboxylase 1 and 2 (ACC1 and ACC2). Key findings include the discovery of an initial lead that was modestly potent and subsequent medicinal chemistry optimization with a focus on lipophilic efficiency (LipE) to balance overall druglike properties. Free-Wilson methodology provided a clear breakdown of the contributions of specific structural elements to the overall LipE, a rationale for prioritization of virtual compounds for synthesis, and a highly successful prediction of the LipE of the resulting analogues. Further preclinical assays, including in vivo malonyl-CoA reduction in both rat liver (ACC1) and rat muscle (ACC2), identified an advanced analogue that progressed to regulatory toxicity studies.


Bioorganic & Medicinal Chemistry Letters | 2010

Structure-based drug design enables conversion of a DFG-in binding CSF-1R kinase inhibitor to a DFG-out binding mode.

Marvin Jay Meyers; Matthew James Pelc; Satwik Kamtekar; Jacqueline E. Day; Gennadiy I. Poda; Molly K. Hall; Marshall L. Michener; Beverly A. Reitz; Karl J. Mathis; Betsy S. Pierce; Mihir D. Parikh; Deborah A. Mischke; Scott A. Long; John J. Parlow; David R. Anderson; Atli Thorarensen

The work described herein demonstrates the utility of structure-based drug design (SBDD) in shifting the binding mode of an HTS hit from a DFG-in to a DFG-out binding mode resulting in a class of novel potent CSF-1R kinase inhibitors suitable for lead development.


Journal of Medicinal Chemistry | 2017

Discovery of Clinical Candidate 1-{[(2S,3S,4S)-3-Ethyl-4-fluoro-5-oxopyrrolidin-2-yl]methoxy}-7-methoxyisoquinoline-6-carboxamide (PF-06650833), a Potent, Selective Inhibitor of Interleukin-1 Receptor Associated Kinase 4 (IRAK4), by Fragment-Based Drug Design

Katherine L. Lee; Catherine M. Ambler; David R. Anderson; Brian P. Boscoe; Andrea G Bree; Joanne Brodfuehrer; Jeanne S. Chang; Chulho Choi; Seung Won Chung; Kevin J. Curran; Jacqueline E. Day; Christoph Martin Dehnhardt; Ken Dower; Susan E. Drozda; Richard K. Frisbie; Lori Krim Gavrin; Joel Adam Goldberg; Seungil Han; Martin Hegen; David Hepworth; Heidi R. Hope; Satwik Kamtekar; Iain Kilty; Arthur Lee; Lih-Ling Lin; Frank Lovering; Michael Dennis Lowe; John Paul Mathias; Heidi M Morgan; Elizabeth Murphy

Through fragment-based drug design focused on engaging the active site of IRAK4 and leveraging three-dimensional topology in a ligand-efficient manner, a micromolar hit identified from a screen of a Pfizer fragment library was optimized to afford IRAK4 inhibitors with nanomolar potency in cellular assays. The medicinal chemistry effort featured the judicious placement of lipophilicity, informed by co-crystal structures with IRAK4 and optimization of ADME properties to deliver clinical candidate PF-06650833 (compound 40). This compound displays a 5-unit increase in lipophilic efficiency from the fragment hit, excellent kinase selectivity, and pharmacokinetic properties suitable for oral administration.


PLOS ONE | 2017

Discovery of PF-06928215 as a high affinity inhibitor of cGAS enabled by a novel fluorescence polarization assay

Justin Hall; Amy Brault; Fabien Vincent; Shawn Weng; Hong Wang; Darren S. Dumlao; Ann Aulabaugh; Dikran Aivazian; Dana Castro; Ming Chen; Jeffrey S. Culp; Ken Dower; Joseph Gardner; Steven J. Hawrylik; Douglas T. Golenbock; David Hepworth; Mark Horn; Lyn H. Jones; Peter Jones; Eicke Latz; Jing Li; Lih-Ling Lin; Wen Lin; David C. Lin; Frank Lovering; Nootaree Niljanskul; Ryan Nistler; Betsy S. Pierce; Olga Plotnikova; Daniel Schmitt

Cyclic GMP-AMP synthase (cGAS) initiates the innate immune system in response to cytosolic dsDNA. After binding and activation from dsDNA, cGAS uses ATP and GTP to synthesize 2′, 3′ -cGAMP (cGAMP), a cyclic dinucleotide second messenger with mixed 2′-5′ and 3′-5′ phosphodiester bonds. Inappropriate stimulation of cGAS has been implicated in autoimmune disease such as systemic lupus erythematosus, thus inhibition of cGAS may be of therapeutic benefit in some diseases; however, the size and polarity of the cGAS active site makes it a challenging target for the development of conventional substrate-competitive inhibitors. We report here the development of a high affinity (KD = 200 nM) inhibitor from a low affinity fragment hit with supporting biochemical and structural data showing these molecules bind to the cGAS active site. We also report a new high throughput cGAS fluorescence polarization (FP)-based assay to enable the rapid identification and optimization of cGAS inhibitors. This FP assay uses Cy5-labelled cGAMP in combination with a novel high affinity monoclonal antibody that specifically recognizes cGAMP with no cross reactivity to cAMP, cGMP, ATP, or GTP. Given its role in the innate immune response, cGAS is a promising therapeutic target for autoinflammatory disease. Our results demonstrate its druggability, provide a high affinity tool compound, and establish a high throughput assay for the identification of next generation cGAS inhibitors.


Bioconjugate Chemistry | 2016

Determination of Antibody–Drug Conjugate Released Payload Species Using Directed in Vitro Assays and Mass Spectrometric Interrogation

Andrew J. Bessire; T. Eric Ballard; Manoj Charati; Justin Cohen; Michael V. Green; My-Hanh Lam; Frank Loganzo; Birte Nolting; Betsy S. Pierce; Sujiet Puthenveetil; Lee R. Roberts; Klaas Schildknegt; Chakrapani Subramanyam

Antibody-drug conjugates (ADC) are currently an active area of research, focused primarily on oncology therapeutics, but also to a limited extent on other areas such as infectious disease. The success of this type of targeted drug delivery is dependent upon many factors, one of which is the performance of the linker in releasing an active drug moiety under the appropriate conditions. As a tool in the development of linker/payload chemistry, we have developed an in vitro method for the identification of payload species released from ADCs in the presence of lysosomal enzymes. This method utilizes commercially available human liver S9 fraction as the source of these enzymes, and this has certain advantages over lysosomal fractions or purified enzymes. This article describes the characterization and performance of this assay with multiple ADCs composed of known and novel linkers and payloads. Additionally, we report the observation of incomplete degradation of mAb protein chains by lysosomal enzymes in vitro, believed to be the first report of this phenomenon involving an ADC therapeutic.


ACS Chemical Biology | 2017

Microfluidic-Enabled Intracellular Delivery of Membrane Impermeable Inhibitors to Study Target Engagement in Human Primary Cells

Jing Li; Bu Wang; Brian Juba; Michael L. Vazquez; Steve W. Kortum; Betsy S. Pierce; Michael Pacheco; Lee R. Roberts; Joseph Walter Strohbach; Lyn H. Jones; Erik C. Hett; Atli Thorarensen; Jean-Baptiste Telliez; Armon Sharei; Mark Edward Bunnage; Jonathan B. Gilbert

Biochemical screening is a major source of lead generation for novel targets. However, during the process of small molecule lead optimization, compounds with excellent biochemical activity may show poor cellular potency, making structure-activity relationships difficult to decipher. This may be due to low membrane permeability of the molecule, resulting in insufficient intracellular drug concentration. The Cell Squeeze platform increases permeability regardless of compound structure by mechanically disrupting the membrane, which can overcome permeability limitations and bridge the gap between biochemical and cellular studies. In this study, we show that poorly permeable Janus kinase (JAK) inhibitors are delivered into primary cells using Cell Squeeze, inhibiting up to 90% of the JAK pathway, while incubation of JAK inhibitors with or without electroporation had no significant effect. We believe this robust intracellular delivery approach could enable more effective lead optimization and deepen our understanding of target engagement by small molecules and functional probes.

Collaboration


Dive into the Betsy S. Pierce's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paolo Orsini

National University of Ireland

View shared research outputs
Top Co-Authors

Avatar

Frank Lovering

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge