Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
Antibiotics (Basel) ; 13(3)2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38534644

ABSTRACT

The global threat of multidrug-resistant Gram-negative bacterial pathogens necessitates the development of new and effective antibiotics. FtsZ is an essential and highly conserved cytoskeletal protein that is an appealing antibacterial target for new antimicrobial therapeutics. However, the effectiveness of FtsZ inhibitors against Gram-negative species has been limited due in part to poor intracellular accumulation. To address this limitation, we have designed a FtsZ inhibitor (RUP4) that incorporates a chlorocatechol siderophore functionality that can chelate ferric iron (Fe3+) and utilizes endogenous siderophore uptake pathways to facilitate entry into Gram-negative pathogens. We show that RUP4 is active against both Klebsiella pneumoniae and Acinetobacter baumannii, with this activity being dependent on direct Fe3+ chelation and enhanced under Fe3+-limiting conditions. Genetic deletion studies in K. pneumoniae reveal that RUP4 gains entry through the FepA and CirA outer membrane transporters and the FhuBC inner membrane transporter. We also show that RUP4 exhibits bactericidal synergy against K. pneumoniae when combined with select antibiotics, with the strongest synergy observed with PBP2-targeting ß-lactams or MreB inhibitors. In the aggregate, our studies indicate that incorporation of Fe3+-chelating moieties into FtsZ inhibitors is an appealing design strategy for enhancing activity against Gram-negative pathogens of global clinical significance.

2.
ACS Chem Biol ; 18(3): 629-642, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36854145

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is a multidrug-resistant (MDR) bacterial pathogen of acute clinical significance. Resistance to current standard-of-care antibiotics, such as vancomycin and linezolid, among nosocomial and community-acquired MRSA clinical isolates is on the rise. This threat to global public health highlights the need to develop new antibiotics for the treatment of MRSA infections. Here, we describe a new benzamide FtsZ inhibitor (TXH9179) with superior antistaphylococcal activity relative to earlier-generation benzamides like PC190723 and TXA707. TXH9179 was found to be 4-fold more potent than TXA707 against a library of 55 methicillin-sensitive S. aureus (MSSA) and MRSA clinical isolates, including MRSA isolates resistant to vancomycin and linezolid. TXH9179 was also associated with a lower frequency of resistance relative to TXA707 in all but one of the MSSA and MRSA isolates examined, with the observed resistance being due to mutations in the ftsZ gene. TXH9179 induced changes in MRSA cell morphology, cell division, and FtsZ localization are fully consistent with its actions as a FtsZ inhibitor. Crystallographic studies demonstrate the direct interaction of TXH9179 with S. aureus FtsZ (SaFtsZ), while delineating the key molecular contacts that drive complex formation. TXH9179 was not associated with any mammalian cytotoxicity, even at a concentration 10-fold greater than that producing antistaphylococcal activity. In serum, the carboxamide prodrug of TXH9179 (TXH1033) is rapidly hydrolyzed to TXH9179 by serum acetylcholinesterases. Significantly, both intravenously and orally administered TXH1033 exhibited enhanced in vivo efficacy relative to the carboxamide prodrug of TXA707 (TXA709) in treating a mouse model of systemic (peritonitis) MRSA infection. Viewed as a whole, our results highlight TXH9179 as a promising new benzamide FtsZ inhibitor worthy of further development.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Prodrugs , Staphylococcal Infections , Animals , Mice , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/chemistry , Benzamides/pharmacology , Benzamides/therapeutic use , Cytoskeletal Proteins/chemistry , Linezolid/pharmacology , Linezolid/therapeutic use , Mammals , Methicillin/pharmacology , Methicillin/therapeutic use , Microbial Sensitivity Tests , Prodrugs/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus , Vancomycin/pharmacology
3.
Antibiotics (Basel) ; 11(5)2022 May 20.
Article in English | MEDLINE | ID: mdl-35625337

ABSTRACT

The emergence of multi-drug-resistant Gram-negative pathogens highlights an urgent clinical need to explore and develop new antibiotics with novel antibacterial targets. MreB is a promising antibacterial target that functions as an essential elongasome protein in most Gram-negative bacterial rods. Here, we describe a third-generation MreB inhibitor (TXH11106) with enhanced bactericidal activity versus the Gram-negative pathogens Escherichia coli, Klebsiella pneumoniae, Acinetobacter baumannii, and Pseudomonas aeruginosa compared to the first- and second-generation compounds A22 and CBR-4830, respectively. Large inocula of these four pathogens are associated with a low frequency of resistance (FOR) to TXH11106. The enhanced bactericidal activity of TXH11106 relative to A22 and CBR-4830 correlates with a correspondingly enhanced capacity to inhibit E. coli MreB ATPase activity via a noncompetitive mechanism. Morphological changes induced by TXH11106 in E. coli, K. pneumoniae, A. baumannii, and P. aeruginosa provide further evidence supporting MreB as the bactericidal target of the compound. Taken together, our results highlight the potential of TXH11106 as an MreB inhibitor with activity against a broad spectrum of Gram-negative bacterial pathogens of acute clinical importance.

4.
Med Chem Res ; 31(10): 1679-1704, 2022 Oct.
Article in English | MEDLINE | ID: mdl-37077288

ABSTRACT

MreB is a cytoskeleton protein present in rod-shaped bacteria that is both essential for bacterial cell division and highly conserved. Because most Gram (-) bacteria require MreB for cell division, chromosome segregation, cell wall morphogenesis, and cell polarity, it is an attractive target for antibacterial drug discovery. As MreB modulation is not associated with the activity of antibiotics in clinical use, acquired resistance to MreB inhibitors is also unlikely. Compounds, such as A22 and CBR-4830, are known to disrupt MreB function by inhibition of ATPase activity. However, the toxicity of these compounds has hindered efforts to assess the in vivo efficacy of these MreB inhibitors. The present study further examines the structure-activity of analogs related to CBR-4830 as it relates to relative antibiotic activity and improved drug properties. These data reveal that certain analogs have enhanced antibiotic activity. In addition, we evaluated several representative analogs (9, 10, 14, 26, and 31) for their abilities to target purified E. coli MreB (EcMreB) and inhibit its ATPase activity. Except for 14, all these analogs were more potent than CBR-4830 as inhibitors of the ATPase activity of EcMreB with corresponding IC50 values ranging from 6 ± 2 to 29 ± 9 µM.

5.
Med Chem Res ; 31(10): 1705-1715, 2022 Oct.
Article in English | MEDLINE | ID: mdl-37065467

ABSTRACT

Oxacillin is a first-line antibiotic for the treatment of methicillin-sensitive Staphylococcus aureus (MSSA) infections but is ineffective against methicillin-resistant S. aureus (MRSA) due to resistance. Here we present results showing that co-administering oxacillin with the FtsZ-targeting prodrug TXA709 renders oxacillin efficacious against MRSA. The combination of oxacillin and the active product of TXA709 (TXA707) is associated with synergistic bactericidal activity against clinical isolates of MRSA that are resistant to current standard-of-care antibiotics. We show that MRSA cells treated with oxacillin in combination with TXA707 exhibit morphological characteristics and PBP2 mislocalization behavior similar to that exhibited by MSSA cells treated with oxacillin alone. Co-administration with TXA709 renders oxacillin efficacious in mouse models of both systemic and tissue infection with MRSA, with this efficacy being observed at human-equivalent doses of oxacillin well below that recommended for daily adult use. Pharmacokinetic evaluations in mice reveal that co-administration with TXA709 also increases total exposure to oxacillin. Viewed as a whole, our results highlight the clinical potential of repurposing oxacillin to treat MRSA infections through combination with a FtsZ inhibitor.

6.
Sci Rep ; 9(1): 20092, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882782

ABSTRACT

Addressing the growing problem of antibiotic resistance requires the development of new drugs with novel antibacterial targets. FtsZ has been identified as an appealing new target for antibacterial agents. Here, we describe the structure-guided design of a new fluorescent probe (BOFP) in which a BODIPY fluorophore has been conjugated to an oxazole-benzamide FtsZ inhibitor. Crystallographic studies have enabled us to identify the optimal position for tethering the fluorophore that facilitates the high-affinity FtsZ binding of BOFP. Fluorescence anisotropy studies demonstrate that BOFP binds the FtsZ proteins from the Gram-positive pathogens Staphylococcus aureus, Enterococcus faecalis, Enterococcus faecium, Streptococcus pyogenes, Streptococcus agalactiae, and Streptococcus pneumoniae with Kd values of 0.6-4.6 µM. Significantly, BOFP binds the FtsZ proteins from the Gram-negative pathogens Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii with an even higher affinity (Kd = 0.2-0.8 µM). Fluorescence microscopy studies reveal that BOFP can effectively label FtsZ in all the above Gram-positive and Gram-negative pathogens. In addition, BOFP is effective at monitoring the impact of non-fluorescent inhibitors on FtsZ localization in these target pathogens. Viewed as a whole, our results highlight the utility of BOFP as a powerful tool for identifying new broad-spectrum FtsZ inhibitors and understanding their mechanisms of action.


Subject(s)
Bacterial Proteins/metabolism , Cytoskeletal Proteins/metabolism , Fluorescent Dyes/chemistry , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/metabolism , Bacterial Proteins/antagonists & inhibitors , Cytoskeletal Proteins/antagonists & inhibitors , Molecular Structure
8.
Eur J Med Chem ; 178: 30-38, 2019 Sep 15.
Article in English | MEDLINE | ID: mdl-31173969

ABSTRACT

Several studies that have identified agents that potentiate the antimicrobial activity of antibiotics, but there are limited insights into their structure-activity relationships (SAR). The SAR associated with select N-alkylaryl amide derivatives of ornithine was performed to establish those structural features that were associated with potentiation of the antimicrobial activity of clarithromycin against E. coli ATCC 25922. The data indicate that the N-propyl derivative was slightly more active in reducing the effective MIC of clarithromycin against E. coli ATCC 25922. In addition, the S-enantiomer of compound 9 was somewhat more potent than the R-enantiomer in potentiating clarithromycin activity. No significant enhancement in potentiation activity was observed with the conversion of these secondary amides to their N-methyl tertiary amides. Formation of the N-methyl or N,N-dimethyl derivatives of the primary amine of 9 was associated with the loss of potentiation activity. Conversion of this primary amine to a guanidine was also not associated with an increase in potentiation activity. Among the isomeric diamino pentamides, 15 potentiated the antibacterial activity of clarithromycin to the greatest extent. In addition to these amide derivatives, the desoxy derivatives 16 and 18 were the more potent potentiators within this triamine series. The relative location of the primary amines, as indicated by the relative differences in the potentiation observed with 16 compared to 14, appears to be a critical factor in determining potentiation activity. Cell-based membrane permeabilization and efflux inhibition studies in E. coli ATCC 25922 suggest that the potentiation of clarithromycin activity by 16 reflects its ability to inhibit efflux pump activity and to a lesser extent its actions as a permeabilizer of the outer leaflet of the outer cell membrane.


Subject(s)
Amides/pharmacology , Anti-Bacterial Agents/pharmacology , Clarithromycin/pharmacology , Escherichia coli/drug effects , Ornithine/pharmacology , Amides/chemical synthesis , Amides/chemistry , Cell Membrane Permeability/drug effects , Drug Synergism , Membrane Transport Proteins/drug effects , Microbial Sensitivity Tests , Molecular Structure , Ornithine/analogs & derivatives , Ornithine/chemical synthesis , Structure-Activity Relationship
9.
ChemMedChem ; 14(12): 1204-1223, 2019 06 18.
Article in English | MEDLINE | ID: mdl-30983160

ABSTRACT

Seasonal influenza infections are associated with an estimated 250-500 000 deaths annually. Resistance to the antiviral M2 ion-channel inhibitors has largely invalidated their clinical utility. Resistance to neuraminidase inhibitors has also been observed in several influenza A virus (IAV) strains. These data have prompted research on inhibitors that target the cap-snatching endonuclease activity of the polymerase acidic protein (PA). Baloxavir marboxil (Xofluza®), recently approved for clinical use, inhibits cap-snatching endonuclease. Resistance to Xofluza® has been reported in both in vitro systems and in the clinic. An X-ray crystallographic screening campaign of a fragment library targeting IAV endonuclease identified 5-chloro-3-hydroxypyridin-2(1H)-one as a bimetal chelating agent at the active site. We have reported the structure-activity relationships for 3-hydroxypyridin-2(1H)-ones and 3-hydroxyquinolin-2(1H)-ones as endonuclease inhibitors. These studies identified two distinct binding modes associated with inhibition of this enzyme that are influenced by the presence of substituents at the 5- and 6-positions of 3-hydroxypyridin-2(1H)-ones. Herein we report the structure-activity relationships associated with various para-substituted 5-phenyl derivatives of 6-(p-fluorophenyl)-3-hydroxypyridin-2(1H)-ones and the effect of using naphthyl, benzyl, and naphthylmethyl groups as alternatives to the p-fluorophenyl substituent on their activity as endonuclease inhibitors.


Subject(s)
Endonucleases/antagonists & inhibitors , Influenza A virus/drug effects , Influenza A virus/enzymology , Pyridones/chemical synthesis , Pyridones/pharmacology , Animals , Antiviral Agents , Crystallography, X-Ray , Dogs , Dose-Response Relationship, Drug , Endonucleases/metabolism , Enzyme Inhibitors , Madin Darby Canine Kidney Cells , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Pyridones/chemistry , Structure-Activity Relationship
10.
ACS Chem Biol ; 12(7): 1947-1955, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28621933

ABSTRACT

In the effort to combat antibiotic resistance, inhibitors of the essential bacterial protein FtsZ have emerged as a promising new class of compounds with clinical potential. One such FtsZ inhibitor (TXA707) is associated with potent activity against clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA) that are resistant to current standard-of-care antibiotics. However, mutations in S. aureus FtsZ (SaFtsZ) that confer resistance to TXA707 have been observed, with mutations in the Gly196 and Gly193 residues being among the most prevalent. Here, we describe structural studies of an FtsZ inhibitor, TXA6101, which retains activity against MRSA isolates that express either G196S or G193D mutant FtsZ. We present the crystal structures of TXA6101 in complex with both wild-type SaFtsZ and G196S mutant SaFtsZ, as well the crystal structure of TXA707 in complex with wild-type SaFtsZ. Comparison of the three structures reveals a molecular basis for the differential targeting abilities of TXA6101 and TXA707. The greater structural flexibility of TXA6101 relative to TXA707 enables TXA6101 to avoid steric clashes with Ser196 and Asp193. Our structures also demonstrate that the binding of TXA6101 induces previously unobserved conformational rearrangements of SaFtsZ residues in the binding pocket. In aggregate, the structures reported in this work reveal key factors for overcoming drug resistance mutations in SaFtsZ and offer a structural basis for the design of FtsZ inhibitors with enhanced antibacterial potency and reduced susceptibility to mutational resistance.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Drug Resistance/genetics , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Mutation , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Crystallography, X-Ray , Models, Molecular
11.
Article in English | MEDLINE | ID: mdl-28630190

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is a multidrug-resistant pathogen that poses a significant risk to global health today. We have developed a promising new FtsZ-targeting agent (TXA707) with potent activity against MRSA isolates resistant to current standard-of-care antibiotics. We present here results that demonstrate differing extents of synergy between TXA707 and a broad range of ß-lactam antibiotics (including six cephalosporins, two penicillins, and two carbapenems) against MRSA. To explore whether there is a correlation between the extent of synergy and the preferential antibacterial target of each ß-lactam, we determined the binding affinities of the ß-lactam antibiotics for each of the four native penicillin-binding proteins (PBPs) of S. aureus using a fluorescence anisotropy competition assay. A comparison of the resulting PBP binding affinities with our corresponding synergy results reveals that ß-lactams with a high affinity for PBP2 afford the greatest degree of synergy with TXA707 against MRSA. In addition, we present fluorescence and electron microscopy studies that suggest a potential mechanism underlying the synergy between TXA707 and the ß-lactam antibiotics. In this connection, our microscopy results show a disruption of septum formation in TXA707-treated MRSA cells, with a concomitant mislocalization of the PBPs from midcell to nonproductive peripheral sites. Viewed as a whole, our results indicate that PBP2-targeting ß-lactam antibiotics are optimal synergistic partners with FtsZ-targeting agents for use in combination therapy of MRSA infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Penicillin-Binding Proteins/metabolism , beta-Lactams/pharmacology , Methicillin/pharmacology , Methicillin Resistance/drug effects , Methicillin-Resistant Staphylococcus aureus/metabolism , Microbial Sensitivity Tests/methods
12.
Antimicrob Agents Chemother ; 60(7): 4290-6, 2016 07.
Article in English | MEDLINE | ID: mdl-27161635

ABSTRACT

Combination therapy of bacterial infections with synergistic drug partners offers distinct advantages over monotherapy. Among these advantages are (i) a reduction of the drug dose required for efficacy, (ii) a reduced potential for drug-induced toxicity, and (iii) a reduced potential for the emergence of resistance. Here, we describe the synergistic actions of the third-generation oral cephalosporin cefdinir and TXA709, a new, FtsZ-targeting prodrug that we have developed with improved pharmacokinetics and enhanced in vivo efficacy against methicillin-resistant Staphylococcus aureus (MRSA) relative to earlier agents. We show that the active product of TXA709 (TXA707) acts synergistically with cefdinir in vitro against clinical isolates of MRSA, vancomycin-intermediate S. aureus (VISA), vancomycin-resistant S. aureus (VRSA), and linezolid-resistant S. aureus (LRSA). In addition, relative to TXA707 alone, the combination of TXA707 and cefdinir significantly reduces or eliminates the detectable emergence of resistance. We also demonstrate synergy in vivo with oral administration of the prodrug TXA709 and cefdinir in mouse models of both systemic and tissue (thigh) infections with MRSA. This synergy reduces the dose of TXA709 required for efficacy 3-fold. Viewed as a whole, our results highlight the potential of TXA709 and cefdinir as a promising combination for the treatment of drug-resistant staphylococcal infections.


Subject(s)
Cephalosporins/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Prodrugs/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Cefdinir , Cytoskeletal Proteins/metabolism , Drug Synergism , Linezolid/pharmacology , Methicillin/pharmacology , Methicillin Resistance/genetics , Microbial Sensitivity Tests , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Vancomycin/pharmacology , Vancomycin Resistance/genetics
13.
Antimicrob Agents Chemother ; 59(8): 4845-55, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26033735

ABSTRACT

The clinical development of FtsZ-targeting benzamide compounds like PC190723 has been limited by poor drug-like and pharmacokinetic properties. Development of prodrugs of PC190723 (e.g., TXY541) resulted in enhanced pharmaceutical properties, which, in turn, led to improved intravenous efficacy as well as the first demonstration of oral efficacy in vivo against both methicillin-sensitive Staphylococcus aureus (MSSA) and methicillin-resistant S. aureus (MRSA). Despite being efficacious in vivo, TXY541 still suffered from suboptimal pharmacokinetics and the requirement of high efficacious doses. We describe here the design of a new prodrug (TXA709) in which the Cl group on the pyridyl ring has been replaced with a CF3 functionality that is resistant to metabolic attack. As a result of this enhanced metabolic stability, the product of the TXA709 prodrug (TXA707) is associated with improved pharmacokinetic properties (a 6.5-fold-longer half-life and a 3-fold-greater oral bioavailability) and superior in vivo antistaphylococcal efficacy relative to PC190723. We validate FtsZ as the antibacterial target of TXA707 and demonstrate that the compound retains potent bactericidal activity against S. aureus strains resistant to the current standard-of-care drugs vancomycin, daptomycin, and linezolid. These collective properties, coupled with minimal observed toxicity to mammalian cells, establish the prodrug TXA709 as an antistaphylococcal agent worthy of clinical development.


Subject(s)
Bacterial Proteins/metabolism , Benzamides/pharmacology , Benzamides/pharmacokinetics , Cytoskeletal Proteins/metabolism , Methicillin Resistance/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Animals , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Cells, Cultured , Daptomycin/pharmacology , Dogs , Half-Life , Humans , Linezolid/pharmacology , Methicillin/pharmacology , Methicillin-Resistant Staphylococcus aureus/metabolism , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests/methods , Pyridines/pharmacology , Rats , Staphylococcal Infections/drug therapy , Thiazoles/pharmacology , Vancomycin/pharmacology
14.
Int J Pharm ; 476(1-2): 199-204, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25263100

ABSTRACT

TXA497 is representative of a new class of guanidinomethyl biaryl compounds that exhibit potent bactericidal behavior against methicillin-resistant Staphylococcus aureus (MRSA). In this study, we compared the anti-staphylococcal, skin deposition, and skin permeation properties of TXA497 and the topical anti-MRSA antibiotic mupirocin. The results of minimum inhibitory concentration (MIC) assays revealed that TXA497 retains potent activity against MRSA that is highly resistant to mupirocin. Using Franz diffusion cells, compound deposition into human cadaver skin was evaluated, and the results showed the skin deposition of TXA497 to be significantly greater than that of mupirocin. Moreover, unlike mupirocin, TXA497 does not pass through the entire skin layer, suggesting a minimal potential for the systemic absorption of the compound upon topical administration. Additionally, antibacterial concentrations of TXA497 showed no significant toxicity to primary human keratinocytes. Given the rising levels of mupirocin resistance among MRSA populations, our results are significant in that they highlight TXA497 as a potentially useful alternative therapy for treating MRSA skin infections that are resistant to mupirocin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biphenyl Compounds/administration & dosage , Guanidines/administration & dosage , Methicillin-Resistant Staphylococcus aureus/drug effects , Mupirocin/administration & dosage , Administration, Cutaneous , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Biphenyl Compounds/pharmacokinetics , Biphenyl Compounds/pharmacology , Drug Resistance, Bacterial , Guanidines/pharmacokinetics , Guanidines/pharmacology , Humans , In Vitro Techniques , Keratinocytes/metabolism , Male , Microbial Sensitivity Tests , Middle Aged , Mupirocin/pharmacokinetics , Mupirocin/pharmacology , Skin/metabolism , Skin Absorption , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology
15.
J Med Chem ; 57(19): 8086-98, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25225968

ABSTRACT

Seasonal and pandemic influenza outbreaks remain a major human health problem. Inhibition of the endonuclease activity of influenza RNA-dependent RNA polymerase is attractive for the development of new agents for the treatment of influenza infection. Our earlier studies identified a series of 5- and 6-phenyl substituted 3-hydroxypyridin-2(1H)-ones that were effective inhibitors of influenza endonuclease. These agents identified as bimetal chelating ligands binding to the active site of the enzyme. In the present study, several aza analogues of these phenyl substituted 3-hydroxypyridin-2(1H)-one compounds were synthesized and evaluated for their ability to inhibit the endonuclease activity. In contrast to the 4-aza analogue of 6-(4-fluorophenyl)-3-hydroxypyridin-2(1H)-one, the 5-aza analogue (5-hydroxy-2-(4-fluorophenyl)pyrimidin-4(3H)-one) did exhibit significant activity as an endonuclease inhibitor. The 6-aza analogue of 5-(4-fluorophenyl)-3-hydroxypyridin-2(1H)-one (6-(4-fluorophenyl)-4-hydroxypyridazin-3(2H)-one) also retained modest activity as an inhibitor. Several varied 6-phenyl-4-hydroxypyridazin-3(2H)-ones and 2-phenyl-5-hydroxypyrimidin-4(3H)-ones were synthesized and evaluated as endonuclease inhibitors. The SAR observed for these aza analogues are consistent with those previously observed with various phenyl substituted 3-hydroxypyridin-2(1H)-ones.


Subject(s)
Antiviral Agents/chemical synthesis , Aza Compounds/chemical synthesis , Endonucleases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Influenza A virus/enzymology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Antiviral Agents/pharmacology , Aza Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Influenza A virus/drug effects , Pyridazines/chemical synthesis , Pyridazines/pharmacology , Structure-Activity Relationship
16.
Biochem Pharmacol ; 89(3): 321-8, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24637241

ABSTRACT

Infections caused by Gram-negative bacterial pathogens are often difficult to treat, with the emergence of multidrug-resistant strains further restricting clinical treatment options. As a result, there is an acute need for the development of new therapeutic agents active against Gram-negative bacteria. The bacterial protein FtsZ has recently been demonstrated to be a viable antibacterial target for treating infections caused by the Gram-positive bacteria Staphylococcus aureus in mouse model systems. Here, we investigate whether an FtsZ-directed prodrug (TXY436) that is effective against S. aureus can also target Gram-negative bacteria, such as Escherichia coli. We find that the conversion product of TXY436 (PC190723) can bind E. coli FtsZ and inhibit its polymerization/bundling in vitro. However, PC190723 is intrinsically inactive against wild-type E. coli, with this inactivity being derived from the actions of the efflux pump AcrAB. Mutations in E. coli AcrAB render the mutant bacteria susceptible to TXY436. We further show that chemical inhibition of AcrAB in E. coli, as well as its homologs in Klebsiella pneumoniae and Acinetobacter baumannii, confers all three Gram-negative pathogens with susceptibility to TXY436. We demonstrate that the activity of TXY436 against E. coli and K. pneumoniae is bactericidal in nature. Evidence for FtsZ-targeting and inhibition of cell division in Gram-negative bacteria by TXY436 is provided by the induction of a characteristic filamentous morphology when the efflux pump has been inhibited as well as by the lack of functional Z-rings upon TXY436 treatment.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Benzamides/pharmacology , Cytoskeletal Proteins/metabolism , Escherichia coli/drug effects , Prodrugs , Pyridines/pharmacology , Thiazoles/pharmacology , rho GTP-Binding Proteins/antagonists & inhibitors , Animals , Anti-Bacterial Agents/chemistry , Benzamides/chemistry , Gene Expression Regulation, Bacterial/drug effects , Klebsiella pneumoniae/drug effects , Microbial Sensitivity Tests , Molecular Structure , Pyridines/chemistry , Thiazoles/chemistry , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
17.
Biochem Pharmacol ; 86(12): 1699-707, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24148278

ABSTRACT

The benzamide derivative PC190723 was among the first of a promising new class of FtsZ-directed antibacterial agents to be identified that exhibit potent antistaphylococcal activity. However, the compound is associated with poor drug-like properties. As part of an ongoing effort to develop FtsZ-targeting antibacterial agents with increased potential for clinical utility, we describe herein the pharmacodynamics, pharmacokinetics, in vivo antistaphylococcal efficacy, and mammalian cytotoxicity of TXY541, a novel 1-methylpiperidine-4-carboxamide prodrug of PC190723. TXY541 was found to be 143-times more soluble than PC190723 in an aqueous acidic vehicle (10mM citrate, pH 2.6) suitable for both oral and intravenous in vivo administration. In staphylococcal growth media, TXY541 converts to PC190723 with a half-life of approximately 8h. In 100% mouse serum, the TXY541-to-PC190723 conversion was much more rapid (with a half-life of approximately 3min), suggesting that the conversion of the prodrug in serum is predominantly enzyme-catalyzed. Pharmacokinetic analysis of both orally and intravenously administered TXY541 in mice yielded a half-life for the PC190723 conversion product of 0.56h and an oral bioavailability of 29.6%. Whether administered orally or intravenously, TXY541 was found to be efficacious in vivo in mouse models of systemic infection with both methicillin-sensitive and methicillin-resistant S. aureus. Toxicological assessment of TXY541 against mammalian cells revealed minimal detectable cytotoxicity. The results presented here highlight TXY541 as a potential therapeutic agent that warrants further pre-clinical development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Heterocyclic Compounds, 2-Ring/pharmacology , Imides/pharmacology , Prodrugs/pharmacology , Pyridines/pharmacology , Staphylococcus/drug effects , Thiazoles/pharmacology , Animals , Anti-Bacterial Agents/pharmacokinetics , Female , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Imides/pharmacokinetics , Mice , Microbial Sensitivity Tests , Prodrugs/pharmacokinetics , Pyridines/pharmacokinetics , Thiazoles/pharmacokinetics
18.
Molecules ; 18(10): 11938-63, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24077174

ABSTRACT

Pyridyl polyoxazoles are 24-membered macrocyclic lactams comprised of a pyridine, four oxazoles and a phenyl ring. A derivative having a 2-(dimethylamino)ethyl chain attached to the 5-position of the phenyl ring was recently identified as a selective G-quadruplex stabilizer with excellent cytotoxic activity, and good in vivo anticancer activity against a human breast cancer xenograft in mice. Here we detail the synthesis of eight new dimethylamino-substituted pyridyl polyoxazoles in which the point of attachment to the macrocycle, as well as the distance between the amine and the macrocycle are varied. Each compound was evaluated for selective G-quadruplex stabilization and cytotoxic activity. The more active analogs have the amine either directly attached to, or separated from the phenyl ring by two methylene groups. There is a correlation between those macrocycles that are effective ligands for the stabilization of G-quadruplex DNA (DT(tran) 15.5-24.6 °C) and cytotoxicity as observed in the human tumor cell lines, RPMI 8402 (IC50 0.06-0.50 µM) and KB3-1 (IC50 0.03-0.07 µM). These are highly selective G-quadruplex stabilizers, which should prove especially useful for evaluating both in vitro and in vivo mechanism(s) of biological activity associated with G-quaqdruplex ligands.


Subject(s)
Antineoplastic Agents/chemical synthesis , G-Quadruplexes , Oxazoles/chemical synthesis , Pyridines/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cyclization , DNA/chemistry , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , Macrocyclic Compounds/chemical synthesis , Oxazoles/pharmacology , Pyridines/pharmacology , Structure-Activity Relationship
19.
Antimicrob Agents Chemother ; 57(12): 5860-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24041882

ABSTRACT

The bacterial cell division protein FtsZ represents a novel antibiotic target that has yet to be exploited clinically. The benzamide PC190723 was among the first FtsZ-targeting compounds to exhibit in vivo efficacy in a murine infection model system. Despite its initial promise, the poor formulation properties of the compound have limited its potential for clinical development. We describe here the development of an N-Mannich base derivative of PC190723 with enhanced drug-like properties and oral in vivo efficacy. The N-Mannich base derivative (TXY436) is ∼100-fold more soluble than PC190723 in an acidic aqueous vehicle (10 mM citrate, pH 2.6) suitable for oral in vivo administration. At physiological pH (7.4), TXY436 acts as a prodrug, converting to PC190723 with a conversion half-life of 18.2 ± 1.6 min. Pharmacokinetic analysis following intravenous administration of TXY436 into mice yielded elimination half-lives of 0.26 and 0.96 h for the TXY436 prodrug and its PC190723 product, respectively. In addition, TXY436 was found to be orally bioavailable and associated with significant extravascular distribution. Using a mouse model of systemic infection with methicillin-sensitive Staphylococcus aureus or methicillin-resistant S. aureus, we show that TXY436 is efficacious in vivo upon oral administration. In contrast, the oral administration of PC190723 was not efficacious. Mammalian cytotoxicity studies of TXY436 using Vero cells revealed an absence of toxicity up to compound concentrations at least 64 times greater than those associated with antistaphylococcal activity. These collective properties make TXY436 a worthy candidate for further investigation as a clinically useful agent for the treatment of staphylococcal infections.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Bacterial Proteins/antagonists & inhibitors , Benzamides/pharmacokinetics , Cytoskeletal Proteins/antagonists & inhibitors , Methicillin-Resistant Staphylococcus aureus/drug effects , Prodrugs/pharmacokinetics , Pyridines/pharmacokinetics , Staphylococcal Infections/drug therapy , Thiazoles/pharmacokinetics , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Benzamides/metabolism , Benzamides/pharmacology , Biological Availability , Biotransformation , Chlorocebus aethiops , Cytoskeletal Proteins/metabolism , Female , Half-Life , Male , Methicillin-Resistant Staphylococcus aureus/growth & development , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Prodrugs/metabolism , Prodrugs/pharmacology , Pyridines/metabolism , Pyridines/pharmacology , Staphylococcal Infections/microbiology , Thiazoles/metabolism , Thiazoles/pharmacology , Vero Cells
20.
Bioorg Med Chem ; 21(21): 6435-46, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24055080

ABSTRACT

Inhibition of the endonuclease activity of influenza RNA-dependent RNA polymerase is recognized as an attractive target for the development of new agents for the treatment of influenza infection. Our earlier study employing small molecule fragment screening using a high-resolution crystal form of pandemic 2009 H1N1 influenza A endonuclease domain (PAN) resulted in the identification of 5-chloro-3-hydroxypyridin-2(1H)-one as a bimetal chelating ligand at the active site of the enzyme. In the present study, several phenyl substituted 3-hydroxypyridin-2(1H)-one compounds were synthesized and evaluated for their ability to inhibit the endonuclease activity as measured by a high-throughput fluorescence assay. Two of the more potent compounds in this series, 16 and 18, had IC50 values of 11 and 23nM in the enzymatic assay, respectively. Crystal structures revealed that these compounds had distinct binding modes that chelate the two active site metal ions (M1 and M2) using only two chelating groups. The SAR and the binding mode of these 3-hydroxypyridin-2-ones provide a basis for developing a new class of anti-influenza drugs.


Subject(s)
Endonucleases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Influenza A Virus, H1N1 Subtype/enzymology , Pyridones/chemistry , Binding Sites , Catalytic Domain , Cell Survival/drug effects , Crystallography, X-Ray , Endonucleases/genetics , Endonucleases/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/toxicity , HEK293 Cells , Humans , Protein Binding , Pyridones/chemical synthesis , Pyridones/toxicity , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...