Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Antimicrob Chemother ; 79(4): 875-882, 2024 04 02.
Article in English | MEDLINE | ID: mdl-38394463

ABSTRACT

BACKGROUND: Non-tuberculous mycobacterial pulmonary disease (NTM-PD) is increasing worldwide, with Mycobacterium avium complex (MAC) and Mycobacterium abscessus as the predominant pathogens. Current treatments are poorly tolerated and modestly effective, highlighting the need for new treatments. SPR719, the active moiety of the benzimidazole prodrug SPR720, inhibits the ATPase subunits of DNA gyrase B, a target not exploited by current antibiotics, and therefore, no cross-resistance is expected with standard-of-care (SOC) agents. OBJECTIVES: To evaluate the in vitro activity of SPR719 against MAC and M. abscessus clinical isolates, including those resistant to SOC agents, and in vivo efficacy of SPR720 in murine non-tuberculous mycobacteria (NTM) pulmonary infection models. METHODS: NTM isolates were tested for susceptibility to SPR719. Chronic C3HeB/FeJ and severe combined immunodeficient murine models of pulmonary infection were used to assess efficacy of SPR720 against MAC and M. abscessus, respectively. RESULTS: SPR719 was active against MAC (MIC90, 2 mg/L) and M. abscessus (MIC90, 4 mg/L) clinical isolates. Efficacy of SPR720 was demonstrated against MAC pulmonary infection, both as a monotherapy and in combination with SOC agents. SPR720 monotherapy exhibited dose-dependent reduction in bacterial burden, with the largest reduction observed when combined with clarithromycin and ethambutol. Efficacy of SPR720 was also demonstrated against M. abscessus pulmonary infection where monotherapy exhibited a dose-dependent reduction in bacterial burden with further reductions detected when combined with SOC agents. CONCLUSIONS: In vitro activity of SPR720 against common NTM pathogens and efficacy in murine infections warrant the continued clinical evaluation of SPR720 as a new oral option for the treatment of NTM-PD.


Subject(s)
Lung Diseases , Mycobacterium Infections, Nontuberculous , Pneumonia , Humans , Animals , Mice , Nontuberculous Mycobacteria , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Disease Models, Animal , Mycobacterium avium Complex , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Lung Diseases/drug therapy , Pneumonia/drug therapy
2.
ACS Infect Dis ; 6(6): 1323-1331, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32183511

ABSTRACT

The prospect of ever increasing antibiotic resistance eroding currently available treatment options for bacterial infections underscores the need to continue to identify new antibiotics, preferably those that act on novel targets or with novel mechanisms of action. Bacterial gyrase B subunit (GyrB), an essential component of bacterial gyrase required for successful DNA replication, represents such a target. We describe recent examples of GyrB inhibitors and point out their potential utility for treatment of mycobacterial diseases caused by Mycobacterium tuberculosis (TB) and non-tuberculous mycobacteria (NTM). Current therapeutic options for these diseases are often suboptimal due to resistance to current standard of care antibiotics. A future GyrB inhibitor-based antibiotic could offer a new and effective addition to the armamentarium for treatment of mycobacterial diseases and possibly for infections caused by other bacterial pathogens. One GyrB inhibitor, SPR720, has recently completed a first-in-human clinical trial and is in clinical development for the treatment of NTM and TB infections.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Humans , Nontuberculous Mycobacteria , Tuberculosis/drug therapy
3.
Bioorg Med Chem Lett ; 24(1): 360-6, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24287382

ABSTRACT

In an attempt to identify novel inhibitors of NAD(+)-dependent DNA ligase (LigA) that are not affected by a known resistance mutation in the adenosine binding pocket, a detailed analysis of the binding sites of a variety of bacterial ligases was performed. This analysis revealed several similarities to the adenine binding region of kinases, which enabled a virtual screen of known kinase inhibitors. From this screen, a thienopyridine scaffold was identified that was shown to inhibit bacterial ligase. Further characterization through structure and enzymology revealed the compound was not affected by a previously disclosed resistance mutation in Streptococcus pneumoniae LigA, Leu75Phe. A subsequent medicinal chemistry program identified substitutions that resulted in an inhibitor with moderate activity across various Gram-positive bacterial LigA enzymes.


Subject(s)
DNA Ligases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Streptococcus pneumoniae/enzymology , DNA Ligases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
4.
Bioorg Med Chem Lett ; 22(23): 7019-23, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23099094

ABSTRACT

A previously described aryl sulfonamide series, originally found through HTS, targets GlmU, a bifunctional essential enzyme involved in bacterial cell wall synthesis. Using structure-guided design, the potency of enzyme inhibition was increased in multiple isozymes from different bacterial species. Unsuitable physical properties (low LogD and high molecular weight) of those compounds prevented them from entering the cytoplasm of bacteria and inhibiting cell growth. Further modifications described herein led to compounds that possessed antibacterial activity, which was shown to occur through inhibition of GlmU. The left-hand side amide and the right-hand side sulfonamides were modified such that enzyme inhibitory activity was maintained (IC(50) <0.1 µM against GlmU isozymes from Gram-negative organisms), and the lipophilicity was increased giving compounds with LogD -1 to 3. Antibacterial activity in an efflux-pump deficient mutant of Haemophilus influenzae resulted for compounds such as 13.


Subject(s)
Acetyltransferases/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Enzyme Inhibitors/chemistry , Nucleotidyltransferases/antagonists & inhibitors , Oxazines/chemistry , Sulfonamides/chemistry , Acetyltransferases/metabolism , Amides/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Binding Sites , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Haemophilus influenzae/drug effects , Haemophilus influenzae/enzymology , Microbial Sensitivity Tests , Molecular Docking Simulation , Nucleotidyltransferases/metabolism , Oxazines/chemical synthesis , Oxazines/pharmacology , Protein Structure, Tertiary , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology
5.
Pharm Pat Anal ; 1(5): 601-20, 2012 Nov.
Article in English | MEDLINE | ID: mdl-24236928

ABSTRACT

This review summarizes patent applications from 2010 for small molecules for which there is a claim of antibacterial activity. The primary criterion for inclusion in this analysis was reporting of cellular antibacterial activity data (MICs) for at least one compound. Patent applications are reviewed according to their biological target and antibacterial class. Protein synthesis inhibitors disclosed in this period include inhibitors of the 50S ribosome subunit (oxazolidinones, macrolides/ketolides and pleuromutilins), 30S ribosome subunit (aminoglycosides and tetracyclines) and nonribosomal targets (PDF inhibitors). DNA synthesis inhibitors include inhibitors of GyrA/ParC and GyrB/ParE. Cell envelope disruptors disclosed in 2010 cover both inhibitors of cell-envelope synthesis (LpxC inhibitors, ß-lactams and glycopeptides), as well as membrane disruptors (lipopeptides and polymyxins). Other antibacterial classes covered in this review include rifamycins and antibacterial peptides. Patent applications for compounds aimed at overcoming resistance mechanisms (efflux inhibitors and ß-lactamase inhibitors) are also described.


Subject(s)
Anti-Bacterial Agents , Animals , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Humans , Nucleic Acid Synthesis Inhibitors/pharmacology , Patents as Topic , Protein Synthesis Inhibitors/pharmacology
6.
Bioorg Med Chem Lett ; 22(1): 85-9, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22154350

ABSTRACT

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.


Subject(s)
Adenosine/chemistry , Anti-Bacterial Agents/chemical synthesis , DNA Ligases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , NAD/chemistry , Adenine/chemistry , Administration, Oral , Animals , Anti-Bacterial Agents/chemistry , Biological Availability , Chromatography, Liquid/methods , DNA Ligases/chemistry , Dogs , Drug Design , Drug Evaluation, Preclinical/methods , Fluorine/chemistry , Inhibitory Concentration 50 , Mass Spectrometry/methods , Models, Chemical , Rats
7.
J Biol Chem ; 286(47): 40734-42, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-21984832

ABSTRACT

GlmU is a bifunctional enzyme that is essential for bacterial growth, converting D-glucosamine 1-phosphate into UDP-GlcNAc via acetylation and subsequent uridyl transfer. A biochemical screen of AstraZeneca's compound library using GlmU of Escherichia coli identified novel sulfonamide inhibitors of the acetyltransferase reaction. Steady-state kinetics, ligand-observe NMR, isothermal titration calorimetry, and x-ray crystallography showed that the inhibitors were competitive with acetyl-CoA substrate. Iterative chemistry efforts improved biochemical potency against gram-negative isozymes 300-fold and afforded antimicrobial activity against a strain of Haemophilus influenzae lacking its major efflux pump. Inhibition of precursor incorporation into bacterial macromolecules was consistent with the antimicrobial activity being caused by disruption of peptidoglycan and fatty acid biosyntheses. Isolation and characterization of two different resistant mutant strains identified the GlmU acetyltransferase domain as the molecular target. These data, along with x-ray co-crystal structures, confirmed the binding mode of the inhibitors and explained their relative lack of potency against gram-positive GlmU isozymes. This is the first example of antimicrobial compounds mediating their growth inhibitory effects specifically via GlmU.


Subject(s)
Acetyltransferases/antagonists & inhibitors , Acetyltransferases/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Haemophilus influenzae/drug effects , Haemophilus influenzae/enzymology , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Sulfonamides/pharmacology , Acetyl Coenzyme A/metabolism , Acetyltransferases/chemistry , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Binding, Competitive , Enzyme Inhibitors/pharmacology , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Inhibitory Concentration 50 , Models, Molecular , Molecular Sequence Data , Multienzyme Complexes/chemistry , Protein Multimerization , Protein Structure, Quaternary , Reproducibility of Results
8.
Bioorg Med Chem Lett ; 21(15): 4556-60, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21719282

ABSTRACT

Optimization of adenosine analog inhibitors of bacterial NAD(+)-dependent DNA ligase is discussed. Antibacterial activity against Streptococcus pneumoniae and Staphylococcus aureus was improved by modification of the 2-position substituent on the adenine ring and 3'- and 5'-substituents on the ribose. Compounds with logD values 1.5-2.5 maximized potency and maintained drug-like physical properties.


Subject(s)
Anti-Bacterial Agents/chemistry , DNA Ligases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Adenosine/analogs & derivatives , Adenosine/chemical synthesis , Adenosine/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Binding Sites , Crystallography, X-Ray , DNA Ligases/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Microbial Sensitivity Tests , NAD/metabolism , Protein Structure, Tertiary , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects
9.
Antimicrob Agents Chemother ; 55(3): 1088-96, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21189350

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , DNA Ligases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Animals , Female , Humans , Mice , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...