Network


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

Hotspot


Dive into the research topics where Joseph V. Newman is active.

Publication


Featured researches published by Joseph V. Newman.


Journal of Medicinal Chemistry | 2011

Novel N-linked aminopiperidine inhibitors of bacterial topoisomerase type II: broad-spectrum antibacterial agents with reduced hERG activity.

Folkert Reck; Richard A. Alm; Patrick Brassil; Joseph V. Newman; Boudewijn Dejonge; Charles J. Eyermann; Gloria Anne Breault; John N. Breen; Janelle Comita-Prevoir; Mark T. D. Cronin; Hajnalka Davis; David E. Ehmann; Vincent Galullo; Bolin Geng; Tyler Grebe; Marshall Morningstar; Phil Walker; Barry Hayter; Stewart L. Fisher

Novel non-fluoroquinolone inhibitors of bacterial type II topoisomerases (DNA gyrase and topoisomerase IV) are of interest for the development of new antibacterial agents that are not impacted by target-mediated cross-resistance with fluoroquinolones. Aminopiperidines that have a bicyclic aromatic moiety linked through a carbon to an ethyl bridge, such as 1, generally show potent broad-spectrum antibacterial activity, including quinolone-resistant isolates, but suffer from potent hERG inhibition (IC(50)= 3 μM for 1). We now disclose the finding that new analogues of 1 with an N-linked cyclic amide moiety attached to the ethyl bridge, such as 24m, retain the broad-spectrum antibacterial activity of 1 but show significantly less hERG inhibition (IC(50)= 31 μM for 24m) and higher free fraction than 1. One optimized analogue, compound 24l, showed moderate clearance in the dog and promising efficacy against Staphylococcus aureus in a mouse thigh infection model.


Antimicrobial Agents and Chemotherapy | 2011

Novel Bacterial NAD+-Dependent DNA Ligase Inhibitors with Broad-Spectrum Activity and Antibacterial Efficacy In Vivo

Scott D. Mills; Ann E. Eakin; Ed T. Buurman; Joseph V. Newman; Ning Gao; Hoan Huynh; Kenneth D. Johnson; Sushmita D. Lahiri; Adam B. Shapiro; Grant K. Walkup; Wei Yang; Suzanne S. Stokes

ABSTRACT DNA ligases are indispensable enzymes playing a critical role in DNA replication, recombination, and repair in all living organisms. Bacterial NAD+-dependent DNA ligase (LigA) was evaluated for its potential as a broad-spectrum antibacterial target. A novel class of substituted adenosine analogs was discovered by target-based high-throughput screening (HTS), and these compounds were optimized to render them more effective and selective inhibitors of LigA. The adenosine analogs inhibited the LigA activities of Escherichia coli, Haemophilus influenzae, Mycoplasma pneumoniae, Streptococcus pneumoniae, and Staphylococcus aureus, with inhibitory activities in the nanomolar range. They were selective for bacterial NAD+-dependent DNA ligases, showing no inhibitory activity against ATP-dependent human DNA ligase 1 or bacteriophage T4 ligase. Enzyme kinetic measurements demonstrated that the compounds bind competitively with NAD+. X-ray crystallography demonstrated that the adenosine analogs bind in the AMP-binding pocket of the LigA adenylation domain. Antibacterial activity was observed against pathogenic Gram-positive and atypical bacteria, such as S. aureus, S. pneumoniae, Streptococcus pyogenes, and M. pneumoniae, as well as against Gram-negative pathogens, such as H. influenzae and Moraxella catarrhalis. The mode of action was verified using recombinant strains with altered LigA expression, an Okazaki fragment accumulation assay, and the isolation of resistant strains with ligA mutations. In vivo efficacy was demonstrated in a murine S. aureus thigh infection model and a murine S. pneumoniae lung infection model. Treatment with the adenosine analogs reduced the bacterial burden (expressed in CFU) in the corresponding infected organ tissue as much as 1,000-fold, thus validating LigA as a target for antibacterial therapy.


Journal of Medicinal Chemistry | 2012

Novel N-linked aminopiperidine inhibitors of bacterial topoisomerase type II with reduced pK(a): antibacterial agents with an improved safety profile.

Folkert Reck; Richard A. Alm; Patrick Brassil; Joseph V. Newman; Paul Ciaccio; John McNulty; Herbert Barthlow; Kosalaram Goteti; John N. Breen; Janelle Comita-Prevoir; Mark T. D. Cronin; David E. Ehmann; Bolin Geng; Andrew A. Godfrey; Stewart L. Fisher

Novel non-fluoroquinolone inhibitors of bacterial type II topoisomerases (DNA gyrase and topoisomerase IV) are of interest for the development of new antibacterial agents that are not impacted by target-mediated cross-resistance with fluoroquinolones. N-Linked amino piperidines, such as 7a, generally show potent antibacterial activity, including against quinolone-resistant isolates, but suffer from hERG inhibition (IC(50) = 44 μM for 7a) and QT prolongation in vivo. We now disclose the finding that new analogues of 7a with reduced pK(a) due to substitution with an electron-withdrawing substituent in the piperidine moiety, such as R,S-7c, retained the Gram-positive activity of 7a but showed significantly less hERG inhibition (IC(50) = 233 μM for R,S-7c). This compound exhibited moderate clearance in dog, promising efficacy against a MRSA strain in a mouse infection model, and an improved in vivo QT profile as measured in a guinea pig in vivo model. As a result of its promising activity, R,S-7c was advanced into phase I clinical studies.


Scientific Reports | 2015

Responding to the challenge of untreatable gonorrhea: ETX0914, a first-in-class agent with a distinct mechanism-of-action against bacterial Type II topoisomerases.

Gregory S. Basarab; Gunther Kern; John McNulty; John P. Mueller; Kenneth Lawrence; Karthick Vishwanathan; Richard A. Alm; Kevin Barvian; Peter Doig; Vincent Galullo; Humphrey Gardner; Madhusudhan Gowravaram; Michael D. Huband; Amy Kimzey; Marshall Morningstar; Amy Kutschke; Sushmita D. Lahiri; Manos Perros; Renu Singh; Virna J. A. Schuck; Ruben Tommasi; Grant K. Walkup; Joseph V. Newman

With the diminishing effectiveness of current antibacterial therapies, it is critically important to discover agents that operate by a mechanism that circumvents existing resistance. ETX0914, the first of a new class of antibacterial agent targeted for the treatment of gonorrhea, operates by a novel mode-of-inhibition against bacterial type II topoisomerases. Incorporating an oxazolidinone on the scaffold mitigated toxicological issues often seen with topoisomerase inhibitors. Organisms resistant to other topoisomerase inhibitors were not cross-resistant with ETX0914 nor were spontaneous resistant mutants to ETX0914 cross-resistant with other topoisomerase inhibitor classes, including the widely used fluoroquinolone class. Preclinical evaluation of ETX0914 pharmacokinetics and pharmacodynamics showed distribution into vascular tissues and efficacy in a murine Staphylococcus aureus infection model that served as a surrogate for predicting efficacious exposures for the treatment of Neisseria gonorrhoeae infections. A wide safety margin to the efficacious exposure in toxicological evaluations supported progression to Phase 1. Dosing ETX0914 in human volunteers showed sufficient exposure and minimal adverse effects to expect a highly efficacious anti-gonorrhea therapy.


ACS Chemical Biology | 2012

In Vivo Validation of Thymidylate Kinase (TMK) with a Rationally Designed, Selective Antibacterial Compound

Thomas A. Keating; Joseph V. Newman; Nelson B. Olivier; Linda G. Otterson; Beth Andrews; P. Ann Boriack-Sjodin; John N. Breen; Peter Doig; Jacques Dumas; Eric Gangl; Oluyinka Green; Satenig Guler; Martin F. Hentemann; Diane Joseph-McCarthy; Sameer Kawatkar; Amy Kutschke; James T. Loch; Andrew R. McKenzie; Selvi Pradeepan; Swati Prasad; Gabriel Martinez-Botella

There is an urgent need for new antibacterials that pinpoint novel targets and thereby avoid existing resistance mechanisms. We have created novel synthetic antibacterials through structure-based drug design that specifically target bacterial thymidylate kinase (TMK), a nucleotide kinase essential in the DNA synthesis pathway. A high-resolution structure shows compound TK-666 binding partly in the thymidine monophosphate substrate site, but also forming new induced-fit interactions that give picomolar affinity. TK-666 has potent, broad-spectrum Gram-positive microbiological activity (including activity against methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococcus), bactericidal action with rapid killing kinetics, excellent target selectivity over the human ortholog, and low resistance rates. We demonstrate in vivo efficacy against S. aureus in a murine infected-thigh model. This work presents the first validation of TMK as a compelling antibacterial target and provides a rationale for pursuing novel clinical candidates for treating Gram-positive infections through TMK.


Nature microbiology | 2017

ETX2514 is a broad-spectrum β-lactamase inhibitor for the treatment of drug-resistant Gram-negative bacteria including Acinetobacter baumannii

Thomas F. Durand-Réville; Satenig Guler; Janelle Comita-Prevoir; Brendan Chen; Neil Bifulco; Hoan Huynh; Sushmita D. Lahiri; Adam B. Shapiro; Sarah M. McLeod; Nicole M. Carter; Samir H. Moussa; Camilo Velez-Vega; Nelson B. Olivier; Robert E. McLaughlin; Ning Gao; Jason Thresher; Tiffany Palmer; Beth Andrews; Robert A. Giacobbe; Joseph V. Newman; David E. Ehmann; Boudewijn L. M. de Jonge; John P. O'Donnell; John P. Mueller; Ruben Tommasi; Alita A. Miller

Multidrug-resistant (MDR) bacterial infections are a serious threat to public health. Among the most alarming resistance trends is the rapid rise in the number and diversity of β-lactamases, enzymes that inactivate β-lactams, a class of antibiotics that has been a therapeutic mainstay for decades. Although several new β-lactamase inhibitors have been approved or are in clinical trials, their spectra of activity do not address MDR pathogens such as Acinetobacter baumannii. This report describes the rational design and characterization of expanded-spectrum serine β-lactamase inhibitors that potently inhibit clinically relevant class A, C and D β-lactamases and penicillin-binding proteins, resulting in intrinsic antibacterial activity against Enterobacteriaceae and restoration of β-lactam activity in a broad range of MDR Gram-negative pathogens. One of the most promising combinations is sulbactam–ETX2514, whose potent antibacterial activity, in vivo efficacy against MDR A. baumannii infections and promising preclinical safety demonstrate its potential to address this significant unmet medical need.


Journal of Medicinal Chemistry | 2012

Discovery of Selective and Potent Inhibitors of Gram-Positive Bacterial Thymidylate Kinase (TMK).

Gabriel Martinez-Botella; John N. Breen; James Duffy; Jacques Dumas; Bolin Geng; Ian K. Gowers; Oluyinka Green; Satenig Guler; Martin F. Hentemann; Felix A. Hernandez-Juan; Diane Joseph-McCarthy; Sameer Kawatkar; Nicholas A. Larsen; Ovadia Lazari; James T. Loch; Jacqueline Macritchie; Andrew R. McKenzie; Joseph V. Newman; Nelson B. Olivier; Linda G. Otterson; Andrew Pate Owens; Jon Read; David W. Sheppard; Thomas A. Keating

Thymidylate kinase (TMK) is an essential enzyme in bacterial DNA synthesis. The deoxythymidine monophosphate (dTMP) substrate binding pocket was targeted in a rational-design, structure-supported effort, yielding a unique series of antibacterial agents showing a novel, induced-fit binding mode. Lead optimization, aided by X-ray crystallography, led to picomolar inhibitors of both Streptococcus pneumoniae and Staphylococcus aureus TMK. MICs < 1 μg/mL were achieved against methicillin-resistant S. aureus (MRSA), S. pneumoniae, and vancomycin-resistant Enterococcus (VRE). Log D adjustments yielded single diastereomers 14 (TK-666) and 46, showing a broad antibacterial spectrum against Gram-positive bacteria and excellent selectivity against the human thymidylate kinase ortholog.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery of bacterial NAD⁺-dependent DNA ligase inhibitors: improvements in clearance of adenosine series.

Suzanne S. Stokes; Madhusudhan Gowravaram; Hoan Huynh; Min Lu; George Mullen; Brendan Chen; Robert Albert; Thomas J. O’Shea; Michael T. Rooney; Haiqing Hu; Joseph V. Newman; Scott D. Mills

Optimization of clearance of adenosine inhibitors of bacterial NAD(+)-dependent DNA ligase is discussed. To reduce Cytochrome P-450-mediated metabolic clearance, many strategies were explored; however, most modifications resulted in compounds with reduced antibacterial activity and/or unchanged total clearance. The alkyl side chains of the 2-cycloalkoxyadenosines were fluorinated, and compounds with moderate antibacterial activity and favorable pharmacokinetic properties in rat and dog were identified.


Bioorganic & Medicinal Chemistry | 2014

Optimization of physicochemical properties and safety profile of novel bacterial topoisomerase type II inhibitors (NBTIs) with activity against Pseudomonas aeruginosa

Folkert Reck; David E. Ehmann; Thomas J. Dougherty; Joseph V. Newman; Sussie Hopkins; Gregory G. Stone; Nikunj Agrawal; Paul Ciaccio; John McNulty; Herbert Barthlow; Jennifer O’Donnell; Kosalaram Goteti; John N. Breen; Janelle Comita-Prevoir; Mark Cornebise; Mark Cronin; Charles J. Eyermann; Bolin Geng; Greg R. Carr; Lakshmipathi Pandarinathan; Xuejun Tang; Andrew Cottone; Liang Zhao; Natascha Bezdenejnih-Snyder

Type II bacterial topoisomerases are well validated targets for antimicrobial chemotherapy. Novel bacterial type II topoisomerase inhibitors (NBTIs) of these targets are of interest for the development of new antibacterial agents that are not impacted by target-mediated cross-resistance with fluoroquinolones. We now disclose the optimization of a class of NBTIs towards Gram-negative pathogens, especially against drug-resistant Pseudomonas aeruginosa. Physicochemical properties (pKa and logD) were optimized for activity against P. aeruginosa and for reduced inhibition of the hERG channel. The optimized analogs 9g and 9i displayed potent antibacterial activity against P. aeruginosa, and a significantly improved hERG profile over previously reported analogs. Compound 9g showed an improved QT profile in in vivo models and lower clearance in rat over earlier compounds. The compounds show promise for the development of new antimicrobial agents against drug-resistant Pseudomonas aeruginosa.


Journal of Medicinal Chemistry | 2014

Antibacterial inhibitors of gram-positive thymidylate kinase: structure-activity relationships and chiral preference of a new hydrophobic binding region.

Sameer Kawatkar; Thomas A. Keating; Nelson B. Olivier; John N. Breen; Oluyinka Green; Satenig Guler; Martin F. Hentemann; James T. Loch; Andrew R. McKenzie; Joseph V. Newman; Linda G. Otterson; Gabriel Martinez-Botella

Thymidylate kinase (TMK), an essential enzyme in bacterial DNA biosynthesis, is an attractive therapeutic target for the development of novel antibacterial agents, and we continue to explore TMK inhibitors with improved potency, protein binding, and pharmacokinetic potential. A structure-guided design approach was employed to exploit a previously unexplored region in Staphylococcus aureus TMK via novel interactions. These efforts produced compound 39, with 3 nM IC50 against S. aureus TMK and 2 μg/mL MIC against methicillin-resistant S. aureus (MRSA). This compound exhibits a striking inverted chiral preference for binding relative to earlier compounds and also has improved physical properties and pharmacokinetics over previously published compounds. An example of this new series was efficacious in a murine S. aureus infection model, suggesting that compounds like 39 are options for further work toward a new Gram-positive antibiotic by maintaining a balance of microbiological potency, low clearance, and low protein binding that can result in lower efficacious doses.

Collaboration


Dive into the Joseph V. Newman'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adam B. Shapiro

Université libre de Bruxelles

View shared research outputs
Researchain Logo
Decentralizing Knowledge