Jeremy T. Starr
Pfizer
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Jeremy T. Starr.
Journal of Medicinal Chemistry | 2014
Mark Edward Flanagan; Joseph A. Abramite; Dennis P. Anderson; Ann Aulabaugh; Upendra P. Dahal; Adam M. Gilbert; Chao Li; Justin Ian Montgomery; Stacey R. Oppenheimer; Tim Ryder; Brandon P. Schuff; Daniel P. Uccello; Gregory S. Walker; Yan Wu; Matthew Frank Brown; Jinshan M. Chen; Matthew Merrill Hayward; Mark C. Noe; R. Scott Obach; Laurence Philippe; Veerabahu Shanmugasundaram; Michael J. Shapiro; Jeremy T. Starr; Justin G. Stroh; Ye Che
Interest in drugs that covalently modify their target is driven by the desire for enhanced efficacy that can result from the silencing of enzymatic activity until protein resynthesis can occur, along with the potential for increased selectivity by targeting uniquely positioned nucleophilic residues in the protein. However, covalent approaches carry additional risk for toxicities or hypersensitivity reactions that can result from covalent modification of unintended targets. Here we describe methods for measuring the reactivity of covalent reactive groups (CRGs) with a biologically relevant nucleophile, glutathione (GSH), along with kinetic data for a broad array of electrophiles. We also describe a computational method for predicting electrophilic reactivity, which taken together can be applied to the prospective design of thiol-reactive covalent inhibitors.
Organic Letters | 2009
Brian S. Gerstenberger; Mark R. Rauckhorst; Jeremy T. Starr
A simple one-pot method for the synthesis of diversely functionalized pyrazoles from aryl nucleophiles, di-tert-butylazodicarboxlate, and 1,3-dicarbonyl or equivalent compounds is presented.
Journal of the American Chemical Society | 2017
Yu Kawamata; Ming Yan; Zhiqing Liu; Deng-Hui Bao; Jinshan Chen; Jeremy T. Starr; Phil S. Baran
A practical electrochemical oxidation of unactivated C–H bonds is presented. This reaction utilizes a simple redox mediator, quinuclidine, with inexpensive carbon and nickel electrodes to selectively functionalize “deep-seated” methylene and methine moieties. The process exhibits a broad scope and good functional group compatibility. The scalability, as illustrated by a 50 g scale oxidation of sclareolide, bodes well for immediate and widespread adoption.
Bioorganic & Medicinal Chemistry Letters | 2009
Jeremy T. Starr; Richard John Sciotti; Debra Hanna; Michael D. Huband; Lisa Mullins; Hongliang Cai; Jeffrey W. Gage; Mandy Lockard; Mark R. Rauckhorst; Robert M. Owen; Manjinder S. Lall; Mark Tomilo; Huifen Chen; Sandra P. McCurdy; Michael R. Barbachyn
Dual inhibitors of bacterial gyrB and parE based on a 5-(2-pyrimidinyl)-imidazo[1,2-a]pyridine template exhibited MICs (microg/mL) of 0.06-64 (Sau), 0.25-64 (MRSA), 0.06-64 (Spy), 0.06-64 (Spn), and 0.03-64 (FQR Spn). Selected examples were efficacious in mouse sepsis and lung infection models at <50mg/kg (PO dosing).
Journal of Medicinal Chemistry | 2014
Jeremy T. Starr; Matthew Frank Brown; Lisa M. Aschenbrenner; Nicole Caspers; Ye Che; Brian S. Gerstenberger; Michael D. Huband; John D. Knafels; M. Megan Lemmon; Chao Li; Sandra P. McCurdy; Eric McElroy; Mark R. Rauckhorst; Andrew P. Tomaras; Jennifer A. Young; Richard P. Zaniewski; Veerabahu Shanmugasundaram; Seungil Han
Multidrug-resistant Gram-negative pathogens are an emerging threat to human health, and addressing this challenge will require development of new antibacterial agents. This can be achieved through an improved molecular understanding of drug-target interactions combined with enhanced delivery of these agents to the site of action. Herein we describe the first application of siderophore receptor-mediated drug uptake of lactivicin analogues as a strategy that enables the development of novel antibacterial agents against clinically relevant Gram-negative bacteria. We report the first crystal structures of several sideromimic conjugated compounds bound to penicillin binding proteins PBP3 and PBP1a from Pseudomonas aeruginosa and characterize the reactivity of lactivicin and β-lactam core structures. Results from drug sensitivity studies with β-lactamase enzymes are presented, as well as a structure-based hypothesis to reduce susceptibility to this enzyme class. Finally, mechanistic studies demonstrating that sideromimic modification alters the drug uptake process are discussed.
Journal of Medicinal Chemistry | 2012
Jun Yong Choi; Mark Stephen Plummer; Jeremy T. Starr; Charlene R. Desbonnet; Holly Soutter; Jeanne Chang; J. Richard Miller; Keith Dillman; Alita A. Miller; William R. Roush
Thymidylate kinase (TMK) is a potential chemotherapeutic target because it is directly involved in the synthesis of an essential component, thymidine triphosphate, in DNA replication. All reported TMK inhibitors are thymidine analogues, which might retard their development as potent therapeutics due to cell permeability and off-target activity against human TMK. A small molecule hit (1, IC(50) = 58 μM), which has reasonable inhibition potency against Pseudomonas aeruginosa TMK (PaTMK), was identified by the analysis of the binding mode of thymidine or TP(5)A in a PaTMK homology model. This hit (1) was cocrystallized with PaTMK, and several potent PaTMK inhibitors (leads, 46, 47, 48, and 56, IC(50) = 100-200 nM) were synthesized using computer-aided design approaches including virtual synthesis/screening, which was used to guide the design of inhibitors. The binding mode of the optimized leads in PaTMK overlaps with that of other bacterial TMKs but not with human TMK, which shares few common features with the bacterial enzymes. Therefore, the optimized TMK inhibitors described here should be useful for the development of antibacterial agents targeting TMK without undesired off-target effects. In addition, an inhibition mechanism associated with the LID loop, which mimics the process of phosphate transfer from ATP to dTMP, was proposed based on X-ray cocrystal structures, homology models, and structure-activity relationship results.
Journal of Medicinal Chemistry | 2013
Matthew Frank Brown; Mark J. Mitton-Fry; Rose Barham; Jeffrey M. Casavant; Brian S. Gerstenberger; Seungil Han; Joel R. Hardink; Thomas M. Harris; Thuy Hoang; Michael D. Huband; Manjinder S. Lall; M. Megan Lemmon; Chao Li; Jian Lin; Sandra P. McCurdy; Eric McElroy; Craig J. McPherson; Eric S. Marr; John P. Mueller; Lisa Mullins; Antonia A. Nikitenko; Mark C. Noe; Joseph Penzien; Mark Stephen Plummer; Brandon P. Schuff; Veerabahu Shanmugasundaram; Jeremy T. Starr; Jianmin Sun; Andrew P. Tomaras; Jennifer A. Young
Herein we describe the structure-aided design and synthesis of a series of pyridone-conjugated monobactam analogues with in vitro antibacterial activity against clinically relevant Gram-negative species including Pseudomonas aeruginosa , Klebsiella pneumoniae , and Escherichia coli . Rat pharmacokinetic studies with compound 17 demonstrate low clearance and low plasma protein binding. In addition, evidence is provided for a number of analogues suggesting that the siderophore receptors PiuA and PirA play a role in drug uptake in P. aeruginosa strain PAO1.
Journal of Medicinal Chemistry | 2013
Thomas V. Magee; Matthew Frank Brown; Jeremy T. Starr; David C. Ackley; Joseph A. Abramite; Jiri Aubrecht; Andrew Butler; Jared L. Crandon; Fadia Dib-Hajj; Mark Edward Flanagan; Karl Granskog; Joel R. Hardink; Michael D. Huband; Rebecca Irvine; Michael Kuhn; Karen L. Leach; Bryan Li; Jian Lin; David R. Luke; Shawn H. MacVane; Alita A. Miller; Sandra P. McCurdy; James M. McKim; David P. Nicolau; Thuy-Trinh Nguyen; Mark C. Noe; John P. O’Donnell; Scott B. Seibel; Yue Shen; Antonia F. Stepan
We report novel polymyxin analogues with improved antibacterial in vitro potency against polymyxin resistant recent clinical isolates of Acinetobacter baumannii and Pseudomonas aeruginosa . In addition, a human renal cell in vitro assay (hRPTEC) was used to inform structure-toxicity relationships and further differentiate analogues. Replacement of the Dab-3 residue with a Dap-3 in combination with a relatively polar 6-oxo-1-phenyl-1,6-dihydropyridine-3-carbonyl side chain as a fatty acyl replacement yielded analogue 5x, which demonstrated an improved in vitro antimicrobial and renal cytotoxicity profiles relative to polymyxin B (PMB). However, in vivo PK/PD comparison of 5x and PMB in a murine neutropenic thigh model against P. aeruginosa strains with matched MICs showed that 5x was inferior to PMB in vivo, suggesting a lack of improved therapeutic index in spite of apparent in vitro advantages.
Angewandte Chemie | 2017
Chao Li; Yu Kawamata; Hugh Nakamura; Julien C. Vantourout; Zhiqing Liu; Qinglong Hou; Deng-Hui Bao; Jeremy T. Starr; Jinshan Chen; Ming Yan; Phil S. Baran
Along with amide bond formation, Suzuki cross-coupling, and reductive amination, the Buchwald-Hartwig-Ullmann-type amination of aryl halides stands as one of the most employed reactions in modern medicinal chemistry. The work herein demonstrates the potential of utilizing electrochemistry to provide a complementary avenue to access such critical bonds using an inexpensive nickel catalyst under mild reaction conditions. Of note is the scalability, functional-group tolerance, rapid rate, and the ability to employ a variety of aryl donors (Ar-Cl, Ar-Br, Ar-I, Ar-OTf), amine types (primary and secondary), and even alternative X-H donors (alcohols and amides).
Organic Letters | 2014
Jinshan Chen; Roberta Properzi; Daniel P. Uccello; Jennifer A. Young; Russell Dushin; Jeremy T. Starr
Reported here are procedures for a one-pot oxidation and rearrangement of propargylamines to synthesize enaminones, with supporting mechanistic studies. Also reported are the extended one-pot syntheses of pyrazoles, including celecoxib and various heterocyclic compounds.