Network


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

Hotspot


Dive into the research topics where Gregory S. Basarab is active.

Publication


Featured researches published by Gregory S. Basarab.


Journal of Medicinal Chemistry | 2013

Fragment-to-Hit-to-Lead Discovery of a Novel Pyridylurea Scaffold of ATP Competitive Dual Targeting Type II Topoisomerase Inhibiting Antibacterial Agents.

Gregory S. Basarab; John Irvin Manchester; Shanta Bist; P.A Boriack-Sjodin; B Dangel; Ruth Illingworth; Brian Sherer; S Sriram; Maria Uria-Nickelsen; Ann E. Eakin

The discovery and optimization of a new class of bacterial topoisomerase (DNA gyrase and topoisomerase IV) inhibitors binding in the ATP domain are described. A fragment molecule, 1-ethyl-3-(2-pyridyl)urea, provided sufficiently potent enzyme inhibition (32 μM) to prompt further analogue work. Acids and acid isosteres were incorporated at the 5-pyridyl position of this fragment, bridging to a key asparagine residue, improving enzyme inhibition, and leading to measurable antibacterial activity. A CF3-thiazole substituent at the 4-pyridyl position improved inhibitory potency due to a favorable lipophilic interaction. Promising antibacterial activity was seen versus the Gram-positive pathogens Staphylococcus aureus and Streptococcus pneumoniae and the Gram-negative pathogens Haemophilus influenzae and Moraxella catarrhalis . Precursor metabolite incorporation and mutant analysis studies support the mode-of-action, blockage of DNA synthesis by dual target topoisomerase inhibition. Compound 35 was efficacious in a mouse S. aureus disease model, where a 4.5-log reduction in colony forming units versus control was demonstrated.


Journal of Medicinal Chemistry | 2014

Optimization of pyrrolamide topoisomerase II inhibitors toward identification of an antibacterial clinical candidate (AZD5099).

Gregory S. Basarab; Pamela Hill; C. Edwin Garner; Ken Hull; Oluyinka Green; Brian Sherer; P. Brian Dangel; John Irvin Manchester; Shanta Bist; Sheila I. Hauck; Fei Zhou; Maria Uria-Nickelsen; Ruth Illingworth; Richard A. Alm; Mike Rooney; Ann E. Eakin

AZD5099 (compound 63) is an antibacterial agent that entered phase 1 clinical trials targeting infections caused by Gram-positive and fastidious Gram-negative bacteria. It was derived from previously reported pyrrolamide antibacterials and a fragment-based approach targeting the ATP binding site of bacterial type II topoisomerases. The program described herein varied a 3-piperidine substituent and incorporated 4-thiazole substituents that form a seven-membered ring intramolecular hydrogen bond with a 5-position carboxylic acid. Improved antibacterial activity and lower in vivo clearances were achieved. The lower clearances were attributed, in part, to reduced recognition by the multidrug resistant transporter Mrp2. Compound 63 showed notable efficacy in a mouse neutropenic Staphylococcus aureus infection model. Resistance frequency versus the drug was low, and reports of clinical resistance due to alteration of the target are few. Hence, 63 could offer a novel treatment for serious issues of resistance to currently used antibacterials.


Journal of Medicinal Chemistry | 2014

Novel DNA gyrase inhibiting spiropyrimidinetriones with a benzisoxazole scaffold: SAR and in vivo characterization.

Gregory S. Basarab; Patrick Brassil; Peter Doig; Vincent Galullo; Howard B. Haimes; Gunther Kern; Amy Kutschke; John McNulty; Virna J. A. Schuck; Gregory G. Stone; Madhusudhan Gowravaram

The compounds described herein with a spirocyclic architecture fused to a benzisoxazole ring represent a new class of antibacterial agents that operate by inhibition of DNA gyrase as corroborated in an enzyme assay and by the inhibition of precursor thymidine into DNA during cell growth. Activity resided in the configurationally lowest energy (2S,4R,4aR) diastereomer. Highly active compounds against Staphylococcus aureus had sufficiently high solubility, high plasma protein free fraction, and favorable pharmacokinetics to suggest that in vivo efficacy could be demonstrated, which was realized with compound (-)-1 in S. aureus mouse infection models. A high drug exposure NOEL on oral dosing in the rat suggested that a high therapeutic margin could be achieved. Importantly, (-)-1 was not cross-resistant with other DNA gyrase inhibitors such as fluoroquinolone and aminocoumarin antibacterials. Hence, this class shows considerable promise for the treatment of infections caused by multidrug resistant bacteria, including S. aureus.


Journal of Biological Chemistry | 2015

Inhibition of Neisseria gonorrhoeae type II Topoisomerases by the Novel Spiropyrimidinetrione AZD0914

Gunther Kern; Tiffany Palmer; David E. Ehmann; Adam B. Shapiro; Beth Andrews; Gregory S. Basarab; Peter Doig; Jun Fan; Ning Gao; Scott D. Mills; John E. Mueller; Shubha Sriram; Jason Thresher; Grant K. Walkup

Background: Inhibition of Neisseria gonorrhoeae type II topoisomerases gyrase and TopoIV by the antibacterial spiropyrimidinetrione AZD0914 was investigated. Results: AZD0914 stabilized the gyrase-DNA complex with double strand DNA cleavage, retaining potency in a fluoroquinolone-resistant mutant, with little inhibition of human type II topoisomerases. Conclusion: AZD0914 displays mechanistic differences from fluoroquinolones. Significance: AZD0914 has the potential to combat drug-resistant gonorrhea. We characterized the inhibition of Neisseria gonorrhoeae type II topoisomerases gyrase and topoisomerase IV by AZD0914 (AZD0914 will be henceforth known as ETX0914 (Entasis Therapeutics)), a novel spiropyrimidinetrione antibacterial compound that is currently in clinical trials for treatment of drug-resistant gonorrhea. AZD0914 has potent bactericidal activity against N. gonorrhoeae, including multidrug-resistant strains and key Gram-positive, fastidious Gram-negative, atypical, and anaerobic bacterial species (Huband, M. D., Bradford, P. A., Otterson, L. G., Basrab, G. S., Giacobe, R. A., Patey, S. A., Kutschke, A. C., Johnstone, M. R., Potter, M. E., Miller, P. F., and Mueller, J. P. (2014) In Vitro Antibacterial Activity of AZD0914: A New Spiropyrimidinetrione DNA Gyrase/Topoisomerase Inhibitor with Potent Activity against Gram-positive, Fastidious Gram-negative, and Atypical Bacteria. Antimicrob. Agents Chemother. 59, 467–474). AZD0914 inhibited DNA biosynthesis preferentially to other macromolecules in Escherichia coli and induced the SOS response to DNA damage in E. coli. AZD0914 stabilized the enzyme-DNA cleaved complex for N. gonorrhoeae gyrase and topoisomerase IV. The potency of AZD0914 for inhibition of supercoiling and the stabilization of cleaved complex by N. gonorrhoeae gyrase increased in a fluoroquinolone-resistant mutant enzyme. When a mutation, conferring mild resistance to AZD0914, was present in the fluoroquinolone-resistant mutant, the potency of ciprofloxacin for inhibition of supercoiling and stabilization of cleaved complex was increased greater than 20-fold. In contrast to ciprofloxacin, religation of the cleaved DNA did not occur in the presence of AZD0914 upon removal of magnesium from the DNA-gyrase-inhibitor complex. AZD0914 had relatively low potency for inhibition of human type II topoisomerases α and β.


Journal of Medicinal Chemistry | 2018

Antimalarial Lead-Optimization Studies on a 2,6-Imidazopyridine Series within a Constrained Chemical Space To Circumvent Atypical Dose–Response Curves against Multidrug Resistant Parasite Strains

Claire Le Manach; Tanya Paquet; Kathryn J. Wicht; Aloysius T. Nchinda; Christel Brunschwig; Mathew Njoroge; Liezl Gibhard; Dale Taylor; Nina Lawrence; Sergio Wittlin; Charles J. Eyermann; Gregory S. Basarab; James Duffy; Paul V. Fish; Leslie J. Street; Kelly Chibale

A lead-optimization program around a 2,6-imidazopyridine scaffold was initiated based on the two early lead compounds, 1 and 2, that were shown to be efficacious in an in vivo humanized Plasmodium falciparum NODscidIL2Rγnull mouse malaria infection model. The observation of atypical dose-response curves when some compounds were tested against multidrug resistant malaria parasite strains guided the optimization process to define a chemical space that led to typical sigmoidal dose-response and complete kill of multidrug resistant parasites. After a structure and property analysis identified such a chemical space, compounds were prepared that displayed suitable activity, ADME, and safety profiles with respect to cytotoxicity and hERG inhibition.


Xenobiotica | 2017

Absorption, distribution, metabolism and elimination of 14C-ETX0914, a novel inhibitor of bacterial type-II topoisomerases in rodents

Jian Guo; Camil Joubran; Ricardo A. Luzietti; Gregory S. Basarab; Scott W. Grimm; Karthick Vishwanathan

Abstract 1. ETX0914 is a novel bacterial topoisomerase inhibitor that has a novel mode-of-inhibition and is in clinical development for the treatment of infections caused by Neisseria gonorrhoeae. 2. The in vitro biotransformation studies of ETX0914 using mouse, rat, dog and human hepatocytes showed moderate intrinsic clearance in mouse and rat and low intrinsic clearance in dog and human. 3. Following intravenous administration of [14C]-ETX0914 in rats, the mean recovery of administered dose in urine, bile and feces was approximately 15%, 55% and 24%, respectively. Unchanged ETX0914 recovered in urine and bile was less than 5% of the dose, indicating that ETX0914 underwent extensive metabolism in rats. Metabolites M1, M2, M4, M6 and M12 detected in both rat and mouse urine samples were not detected in mouse urine when predosed with 1-aminobenzotriazole, indicating that these metabolites were cytochrome P450 mediated products. The major fecal metabolites observed in rats were not formed when ETX0914 was incubated with fresh feces from germ free rats under sterile condition or in incubations with rat intestinal microsome and cytosol, suggesting that most likely ETX0914 was directly excreted into gut lumen where metabolites were formed as intestinal microflora-mediated products. The major sites of metabolism by CYP enzymes were in the morpholine and oxazolidinone rings while it was benzisoxazole reduction with the gut microflora.


Archive | 2017

Four Ways to Skin a Cat: Inhibition of Bacterial Topoisomerases Leading to the Clinic

Gregory S. Basarab

Four classes of antibacterial agents that operate by inhibition of the Type II topoisomerases, DNA gyrase and Topoisomerase IV, have progressed at least through Phase 2 clinical trials. Compounds from each of the four classes are not cross-resistant to one another as determined by analyses with laboratory and clinical resistant bacterial strains. Hence, they are defined herein as sharing a mode of action, in that they inhibit the same targets, but differing in mode of inhibition, in that they obstruct enzyme activity via divergent binding modes. Two of the classes, fluoroquinolones and aminocoumarins, were long ago approved for clinical use, though the use of the latter has been limited. Two newer classes, spiropyrimidinetriones and quinolines, are represented by the advanced drug candidates zoliflodacin and gepotidacin, each featuring a novel scaffold and a distinct binding motif. X-ray crystallography has shown fluoroquinolone and spiropyrimidinetrione binding at DNA cleavage sites of the topoisomerases. However, the two differ by their dependence on [Mg2+] for binding serving in part to explain the lack of cross-resistance. Quinolines bind to DNA-topoisomerase complexes offset from the cleavage sites as ascertained by X-ray crystallography. Novobiocin, the only aminocoumarin to receive regulatory approval, competes with ATP binding at a site quite remote from the DNA-binding domain. As novobiocin has been withdrawn from the clinic, considerable drug discovery efforts have focused on alternative ATP site binders (ATPase inhibitors). With widespread use of fluoroquinolones leading to resistance, the importance of developing novel antibiotics that would not be cross-resistant is clear. Reviewed herein are the current understandings of the respective mechanisms of inhibition and the respective topoisomerase binding modes for the four classes of antibacterials now with clinical proof of concept.


Antimicrobial Agents and Chemotherapy | 2018

UCT943, a next generation Plasmodium falciparum PI4K inhibitor preclinical candidate for the treatment of malaria

Christel Brunschwig; Nina Lawrence; Dale Taylor; Efrem Abay; Mathew Njoroge; Gregory S. Basarab; Claire Le Manach; Tanya Paquet; Diego Gonzàlez Cabrera; Aloysius T. Nchinda; Carmen de Kock; Lubbe Wiesner; Paolo Denti; David Waterson; Benjamin Blasco; Didier Leroy; Michael J. Witty; Cristina Donini; James Duffy; Sergio Wittlin; Karen L. White; Susan A. Charman; María Belén Jiménez-Díaz; Iñigo Angulo-Barturen; Esperanza Herreros; Francisco Javier Gamo; Rosemary Rochford; Dalu Mancama; Theresa L. Coetzer; Mariëtte E. van der Watt


Antimicrobial Agents and Chemotherapy | 2018

Investigating sulfoxide to sulfone conversion as a prodrug strategy for a PI4K inhibitor in a humanized mouse model of malaria

Liezl Gibhard; Mathew Njoroge; Tanya Paquet; Christel Brunschwig; Dale Taylor; Nina Lawrence; Efrem Abay; Sergio Wittlin; Lubbe Wiesner; Leslie J. Street; Kelly Chibale; Gregory S. Basarab


Antimicrobial Agents and Chemotherapy | 2018

Erratum for Brunschwig et al., “UCT943, a Next-Generation Plasmodium falciparum PI4K Inhibitor Preclinical Candidate for the Treatment of Malaria”

Christel Brunschwig; Nina Lawrence; Dale Taylor; Efrem Abay; Mathew Njoroge; Gregory S. Basarab; Claire Le Manach; Tanya Paquet; Diego Gonzàlez Cabrera; Aloysius T. Nchinda; Carmen de Kock; Lubbe Wiesner; Paolo Denti; David Waterson; Benjamin Blasco; Didier Leroy; Michael J. Witty; Cristina Donini; James Duffy; Sergio Wittlin; Karen L. White; Susan A. Charman; María Belén Jiménez-Díaz; Iñigo Angulo-Barturen; Esperanza Herreros; Francisco Javier Gamo; Rosemary Rochford; Dalu Mancama; Theresa L. Coetzer; Mariëtte E. van der Watt

Collaboration


Dive into the Gregory S. Basarab's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dale Taylor

University of Cape Town

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tanya Paquet

University of Cape Town

View shared research outputs
Top Co-Authors

Avatar

Sergio Wittlin

Swiss Tropical and Public Health Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge