Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Antimicrob Agents Chemother ; 67(11): e0079123, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37850746

ABSTRACT

A wide variety of clinically observed single amino acid substitutions in the Ω-loop region have been associated with increased minimum inhibitory concentrations and resistance to ceftazidime (CAZ) and ceftolozane (TOL) in Pseudomonas-derived cephalosporinase and other class C ß-lactamases. Herein, we demonstrate the naturally occurring tyrosine to histidine substitution of amino acid 221 (Y221H) in Pseudomonas-derived cephalosporinase (PDC) enables CAZ and TOL hydrolysis, leading to similar kinetic profiles (k cat = 2.3 ± 0.2 µM and 2.6 ± 0.1 µM, respectively). Mass spectrometry of PDC-3 establishes the formation of stable adducts consistent with the formation of an acyl enzyme complex, while spectra of E219K (a well-characterized, CAZ- and TOL-resistant comparator) and Y221H are consistent with more rapid turnover. Thermal denaturation experiments reveal decreased stability of the variants. Importantly, PDC-3, E219K, and Y221H are all inhibited by avibactam and the boronic acid transition state inhibitors (BATSIs) LP06 and S02030 with nanomolar IC50 values and the BATSIs stabilize all three enzymes. Crystal structures of PDC-3 and Y221H as apo enzymes and complexed with LP06 and S02030 (1.35-2.10 Å resolution) demonstrate ligand-induced conformational changes, including a significant shift in the position of the sidechain of residue 221 in Y221H (as predicted by enhanced sampling well-tempered metadynamics simulations) and extensive hydrogen bonding between the enzymes and BATSIs. The shift of residue 221 leads to the expansion of the active site pocket, and molecular docking suggests substrates orientate differently and make different intermolecular interactions in the enlarged active site compared to the wild-type enzyme.


Subject(s)
Ceftazidime , Cephalosporinase , Ceftazidime/pharmacology , Cephalosporinase/metabolism , Pseudomonas/genetics , Molecular Docking Simulation , beta-Lactamases/metabolism , Protein Engineering , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Azabicyclo Compounds/pharmacology , Pseudomonas aeruginosa/metabolism , Drug Combinations
2.
PLoS One ; 18(6): e0287102, 2023.
Article in English | MEDLINE | ID: mdl-37310985

ABSTRACT

Recently, we reported rifabutin hyper-activity against Acinetobacter baumannii. We sought to characterize if any additional rifamycins (n = 22) would also display hyper-activity when tested in iron-limited media against A. baumannii, K. pneumoniae, and E. coli. MICs were determined against representative clinical isolates using the iron-limited media RPMI-1640. Only rifabutin was hyperactive against A. baumannii.


Subject(s)
Acinetobacter baumannii , Rifamycins , Rifamycins/pharmacology , Escherichia coli , Klebsiella pneumoniae , Rifabutin , Iron/pharmacology , Microbial Sensitivity Tests
3.
J Med Chem ; 66(13): 8510-8525, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37358467

ABSTRACT

Class C Acinetobacter-derived cephalosporinases (ADCs) represent an important target for inhibition in the multidrug-resistant pathogen Acinetobacter baumannii. Many ADC variants have emerged, and characterization of their structural and functional differences is essential. Equally as important is the development of compounds that inhibit all prevalent ADCs despite these differences. The boronic acid transition state inhibitor, MB076, a novel heterocyclic triazole with improved plasma stability, was synthesized and inhibits seven different ADC ß-lactamase variants with Ki values <1 µM. MB076 acted synergistically in combination with multiple cephalosporins to restore susceptibility. ADC variants containing an alanine duplication in the Ω-loop, specifically ADC-33, exhibited increased activity for larger cephalosporins, such as ceftazidime, cefiderocol, and ceftolozane. X-ray crystal structures of ADC variants in this study provide a structural context for substrate profile differences and show that the inhibitor adopts a similar conformation in all ADC variants, despite small changes near their active sites.


Subject(s)
Acinetobacter baumannii , Cephalosporinase , Cephalosporinase/genetics , Cephalosporinase/chemistry , Cephalosporinase/pharmacology , Boronic Acids/pharmacology , Boronic Acids/chemistry , Cephalosporins/pharmacology , beta-Lactamases/genetics , beta-Lactamases/chemistry , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests
4.
Antibiotics (Basel) ; 12(4)2023 Mar 24.
Article in English | MEDLINE | ID: mdl-37107006

ABSTRACT

Acinetobacter baumannii is a Gram-negative organism listed as an urgent threat pathogen by the World Health Organization (WHO). Carbapenem-resistant A. baumannii (CRAB), especially, present therapeutic challenges due to complex mechanisms of resistance to ß-lactams. One of the most important mechanisms is the production of ß-lactamase enzymes capable of hydrolyzing ß-lactam antibiotics. Co-expression of multiple classes of ß-lactamases is present in CRAB; therefore, the design and synthesis of "cross-class" inhibitors is an important strategy to preserve the efficacy of currently available antibiotics. To identify new, nonclassical ß-lactamase inhibitors, we previously identified a sulfonamidomethaneboronic acid CR167 active against Acinetobacter-derived class C ß-lactamases (ADC-7). The compound demonstrated affinity for ADC-7 with a Ki = 160 nM and proved to be able to decrease MIC values of ceftazidime and cefotaxime in different bacterial strains. Herein, we describe the activity of CR167 against other ß-lactamases in A. baumannii: the cefepime-hydrolysing class C extended-spectrum ß-lactamase (ESAC) ADC-33 and the carbapenem-hydrolyzing OXA-24/40 (class D). These investigations demonstrate CR167 as a valuable cross-class (C and D) inhibitor, and the paper describes our attempts to further improve its activity. Five chiral analogues of CR167 were rationally designed and synthesized. The structures of OXA-24/40 and ADC-33 in complex with CR167 and select chiral analogues were obtained. The structure activity relationships (SARs) are highlighted, offering insights into the main determinants for cross-class C/D inhibitors and impetus for novel drug design.

5.
Antimicrob Agents Chemother ; 67(1): e0093022, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36602311

ABSTRACT

Design of novel ß-lactamase inhibitors (BLIs) is one of the currently accepted strategies to combat the threat of cephalosporin and carbapenem resistance in Gram-negative bacteria. Boronic acid transition state inhibitors (BATSIs) are competitive, reversible BLIs that offer promise as novel therapeutic agents. In this study, the activities of two α-amido-ß-triazolylethaneboronic acid transition state inhibitors (S02030 and MB_076) targeting representative KPC (KPC-2) and CTX-M (CTX-M-96, a CTX-M-15-type extended-spectrum ß-lactamase [ESBL]) ß-lactamases were evaluated. The 50% inhibitory concentrations (IC50s) for both inhibitors were measured in the nanomolar range (2 to 135 nM). For S02030, the k2/K for CTX-M-96 (24,000 M-1 s-1) was twice the reported value for KPC-2 (12,000 M-1 s-1); for MB_076, the k2/K values ranged from 1,200 M-1 s-1 (KPC-2) to 3,900 M-1 s-1 (CTX-M-96). Crystal structures of KPC-2 with MB_076 (1.38-Å resolution) and S02030 and the in silico models of CTX-M-96 with these two BATSIs show that interaction in the CTX-M-96-S02030 and CTX-M-96-MB_076 complexes were overall equivalent to that observed for the crystallographic structure of KPC-2-S02030 and KPC-2-MB_076. The tetrahedral interaction surrounding the boron atom from S02030 and MB_076 creates a favorable hydrogen bonding network with S70, S130, N132, N170, and S237. However, the changes from W105 in KPC-2 to Y105 in CTX-M-96 and the missing residue R220 in CTX-M-96 alter the arrangement of the inhibitors in the active site of CTX-M-96, partially explaining the difference in kinetic parameters. The novel BATSI scaffolds studied here advance our understanding of structure-activity relationships (SARs) and illustrate the importance of new approaches to ß-lactamase inhibitor design.


Subject(s)
Triazoles , beta-Lactamases , beta-Lactamases/genetics , beta-Lactamases/chemistry , beta-Lactamase Inhibitors/pharmacology , Boronic Acids/pharmacology , Boronic Acids/chemistry , Penicillins , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests
6.
Front Microbiol ; 12: 720036, 2021.
Article in English | MEDLINE | ID: mdl-34970229

ABSTRACT

The rise of multidrug resistant (MDR) Gram-negative bacteria has accelerated the development of novel inhibitors of class A and C ß-lactamases. Presently, the search for novel compounds with new mechanisms of action is a clinical and scientific priority. To this end, we determined the 2.13-Å resolution crystal structure of S02030, a boronic acid transition state inhibitor (BATSI), bound to MOX-1 ß-lactamase, a plasmid-borne, expanded-spectrum AmpC ß-lactamase (ESAC) and compared this to the previously reported aztreonam (ATM)-bound MOX-1 structure. Superposition of these two complexes shows that S02030 binds in the active-site cavity more deeply than ATM. In contrast, the SO3 interactions and the positional change of the ß-strand amino acids from Lys315 to Asn320 were more prominent in the ATM-bound structure. MICs were performed using a fixed concentration of S02030 (4 µg/ml) as a proof of principle. Microbiological evaluation against a laboratory strain of Escherichia coli expressing MOX-1 revealed that MICs against ceftazidime are reduced from 2.0 to 0.12 µg/ml when S02030 is added at a concentration of 4 µg/ml. The IC50 and K i of S02030 vs. MOX-1 were 1.25 ± 0.34 and 0.56 ± 0.03 µM, respectively. Monobactams such as ATM can serve as informative templates for design of mechanism-based inhibitors such as S02030 against ESAC ß-lactamases.

7.
Org Biomol Chem ; 19(30): 6687-6691, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34286797

ABSTRACT

A straightforward concise synthesis of chiral non-racemic aliphatic α-boryl isocyanides, relay intermediates for boron-based bioactive molecules in multicomponent reactions, is presented. The short synthetic sequence comprises as key steps copper-catalysed asymmetric borylation of imines, simultaneous nitrogen formylation/boron-protecting group interconversion and the final formamide dehydration reaction.

8.
Diagn Microbiol Infect Dis ; 99(2): 115242, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33248392

ABSTRACT

Successful treatment of Acinetobacter baumannii infections require early and appropriate antimicrobial therapy. One of the first steps in this process is understanding which ß-lactamase (bla) alleles are present and in what combinations. Thus, we performed WGS on 98 carbapenem-resistant A. baumannii (CR Ab). In most isolates, an acquired blaOXA carbapenemase was found in addition to the intrinsic blaOXA allele. The most commonly found allele was blaOXA-23 (n = 78/98). In some isolates, blaOXA-23 was found in addition to other carbapenemase alleles: blaOXA-82 (n = 12/78), blaOXA-72 (n = 2/78) and blaOXA-24/40 (n = 1/78). Surprisingly, 20% of isolates carried carbapenemases not routinely assayed for by rapid molecular diagnostic platforms, i.e., blaOXA-82 and blaOXA-172; all had ISAba1 elements. In 8 CR Ab, blaOXA-82 or blaOXA-172 was the only carbapenemase. Both blaOXA-24/40 and its variant blaOXA-72 were each found in 6/98 isolates. The most prevalent ADC variants were blaADC-30 (21%), blaADC-162 (21%), and blaADC-212 (26%). Complete combinations are reported.


Subject(s)
Acinetobacter baumannii/genetics , Bacterial Proteins/genetics , Carbapenems/pharmacology , beta-Lactam Resistance/genetics , beta-Lactamases/genetics , Acinetobacter Infections/microbiology , Acinetobacter baumannii/enzymology , Acinetobacter baumannii/isolation & purification , Genome, Bacterial/genetics , Humans
9.
Antimicrob Agents Chemother ; 64(12)2020 11 17.
Article in English | MEDLINE | ID: mdl-32988830

ABSTRACT

Extended-spectrum class C ß-lactamases have evolved to rapidly inactivate expanded-spectrum cephalosporins, a class of antibiotics designed to be resistant to hydrolysis by ß-lactamase enzymes. To better understand the mechanism by which Acinetobacter-derived cephalosporinase-7 (ADC-7), a chromosomal AmpC enzyme, hydrolyzes these molecules, we determined the X-ray crystal structure of ADC-7 in an acyl-enzyme complex with the cephalosporin ceftazidime (2.40 Å) as well as in complex with a boronic acid transition state analog inhibitor that contains the R1 side chain of ceftazidime (1.67 Å). In the acyl-enzyme complex, the carbonyl oxygen is situated in the oxyanion hole where it makes key stabilizing interactions with the main chain nitrogens of Ser64 and Ser315. The boronic acid O1 hydroxyl group is similarly positioned in this area. Conserved residues Gln120 and Asn152 form hydrogen bonds with the amide group of the R1 side chain in both complexes. These complexes represent two steps in the hydrolysis of expanded-spectrum cephalosporins by ADC-7 and offer insight into the inhibition of ADC-7 by ceftazidime through displacement of the deacylating water molecule as well as blocking its trajectory to the acyl carbonyl carbon. In addition, the transition state analog inhibitor, LP06, was shown to bind with high affinity to ADC-7 (Ki , 50 nM) and was able to restore ceftazidime susceptibility, offering the potential for optimization efforts of this type of inhibitor.


Subject(s)
Acinetobacter , Boronic Acids , Ceftazidime , Cephalosporinase , Anti-Bacterial Agents/pharmacology , Boronic Acids/pharmacology , Ceftazidime/pharmacology , Cephalosporinase/drug effects , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases
10.
ACS Infect Dis ; 6(7): 1965-1975, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32502340

ABSTRACT

Boronic acid transition state inhibitors (BATSIs) are known reversible covalent inhibitors of serine ß-lactamases. The selectivity and high potency of specific BATSIs bearing an amide side chain mimicking the ß-lactam's amide side chain are an established and recognized synthetic strategy. Herein, we describe a new class of BATSIs where the amide group is replaced by a bioisostere triazole; these compounds were designed as molecular probes. To this end, a library of 26 α-triazolylmethaneboronic acids was synthesized and tested against the clinically concerning Acinetobacter-derived cephalosporinase, ADC-7. In steady state analyses, these compounds demonstrated Ki values ranging from 90 nM to 38 µM (±10%). Five compounds were crystallized in complex with ADC-7 ß-lactamase, and all the crystal structures reveal the triazole is in the putative amide binding site, thus confirming the triazole-amide bioisosterism. The easy synthetic access of these new inhibitors as prototype scaffolds allows the insertion of a wide range of chemical groups able to explore the enzyme binding site and provides insights on the importance of specific residues in recognition and catalysis. The best inhibitor identified, compound 6q (Ki 90 nM), places a tolyl group near Arg340, making favorable cation-π interactions. Notably, the structure of 6q does not resemble the natural substrate of the ß-lactamase yet displays a pronounced inhibition activity, in addition to lowering the minimum inhibitory concentration (MIC) of ceftazidime against three bacterial strains expressing class C ß-lactamases. In summary, these observations validate the α-triazolylboronic acids as a promising template for further inhibitor design.


Subject(s)
Acinetobacter baumannii , beta-Lactamase Inhibitors , Acinetobacter baumannii/metabolism , Cephalosporinase/genetics , Cephalosporinase/metabolism , Structure-Activity Relationship , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism
11.
Biomolecules ; 10(5)2020 04 27.
Article in English | MEDLINE | ID: mdl-32349291

ABSTRACT

Boronic acid transition-state analog inhibitors (BATSIs) are partners with ß-lactam antibiotics for the treatment of complex bacterial infections. Herein, microbiological, biochemical, and structural findings on four BATSIs with the FOX-4 cephamycinase, a class C ß-lactamase that rapidly hydrolyzes cefoxitin, are revealed. FOX-4 is an extended-spectrum class C cephalosporinase that demonstrates conformational flexibility when complexed with certain ligands. Like other ß-lactamases of this class, studies on FOX-4 reveal important insights into structure-activity relationships. We show that SM23, a BATSI, shows both remarkable flexibility and affinity, binding similarly to other ß-lactamases, yet retaining an IC50 value < 0.1 µM. Our analyses open up new opportunities for the design of novel transition-state analogs of class C enzymes.


Subject(s)
Anti-Bacterial Agents/chemistry , Cephalothin/analogs & derivatives , Enzyme Inhibitors/chemistry , Escherichia coli Proteins/chemistry , beta-Lactamases/chemistry , Anti-Bacterial Agents/pharmacology , Binding Sites , Boronic Acids/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/metabolism , Molecular Docking Simulation , Protein Binding , beta-Lactamases/metabolism
12.
ChemMedChem ; 15(14): 1283-1288, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32459878

ABSTRACT

Boronic acids are known reversible covalent inhibitors of serine ß-lactamases. The selectivity and high potency of specific boronates bearing an amide side chain that mimics the ß-lactam's amide side chain have been advanced in several studies. Herein, we describe a new class of boronic acids in which the amide group is replaced by a bioisostere triazole. The boronic acids were obtained in a two-step synthesis that relies on the solid and versatile copper-catalyzed azide-alkyne cycloaddition (CuAAC) followed by boronate deprotection. All of the compounds show very good inhibition of the Klebsiella pneumoniae carbapenemase KPC-2, with Ki values ranging from 1 nM to 1 µM, and most of them are able to restore cefepime activity against K. pneumoniae harboring blaKPC-2 . In particular, compound 1 e, bearing a sulfonamide substituted by a thiophene ring, proved to be an excellent KPC-2 inhibitor (Ki =30 nM); it restored cefepime susceptibility in KPC-Kpn cells (MIC=0.5 µg/mL) with values similar to that of vaborbactam (Ki =20 nM, MIC in KPC-Kpn 0.5 µg/mL). Our findings suggest that α-triazolylboronates might represent an effective scaffold for the treatment of KPC-mediated infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Boronic Acids/pharmacology , Klebsiella pneumoniae/drug effects , Triazoles/pharmacology , beta-Lactamase Inhibitors/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Boronic Acids/chemical synthesis , Boronic Acids/chemistry , Dose-Response Relationship, Drug , Klebsiella pneumoniae/enzymology , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , beta-Lactamase Inhibitors/chemical synthesis , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/isolation & purification , beta-Lactamases/metabolism
13.
Front Microbiol ; 11: 35, 2020.
Article in English | MEDLINE | ID: mdl-32117094

ABSTRACT

Pseudomonas aeruginosa is a Gram-negative nosocomial pathogen, often causative agent of severe device-related infections, given its great capacity to form biofilm. P. aeruginosa finely regulates the expression of numerous virulence factors, including biofilm production, by Quorum Sensing (QS), a cell-to-cell communication mechanism used by many bacteria. Selective inhibition of QS-controlled pathogenicity without affecting bacterial growth may represent a novel promising strategy to overcome the well-known and widespread drug resistance of P. aeruginosa. In this study, we investigated the effects of SM23, a boronic acid derivate specifically designed as ß-lactamase inhibitor, on biofilm formation and virulence factors production by P. aeruginosa. Our results indicated that SM23: (1) inhibited biofilm development and production of several virulence factors, such as pyoverdine, elastase, and pyocyanin, without affecting bacterial growth; (2) decreased the levels of 3-oxo-C12-HSL and C4-HSL, two QS-related autoinducer molecules, in line with a dampened lasR/lasI system; (3) failed to bind to bacterial cells that had been preincubated with P. aeruginosa-conditioned medium; and (4) reduced both biofilm formation and pyoverdine production by P. aeruginosa onto endotracheal tubes, as assessed by a new in vitro model closely mimicking clinical settings. Taken together, our results indicate that, besides inhibiting ß-lactamase, SM23 can also act as powerful inhibitor of P. aeruginosa biofilm, suggesting that it may have a potential application in the prevention and treatment of biofilm-associated P. aeruginosa infections.

14.
mBio ; 9(6)2018 12 11.
Article in English | MEDLINE | ID: mdl-30538183

ABSTRACT

Pseudomonas aeruginosa produces a class C ß-lactamase (e.g., PDC-3) that robustly hydrolyzes early generation cephalosporins often at the diffusion limit; therefore, bacteria possessing these ß-lactamases are resistant to many ß-lactam antibiotics. In response to this significant clinical threat, ceftolozane, a 3' aminopyrazolium cephalosporin, was developed. Combined with tazobactam, ceftolozane promised to be effective against multidrug-resistant P. aeruginosa Alarmingly, Ω-loop variants of the PDC ß-lactamase (V213A, G216R, E221K, E221G, and Y223H) were identified in ceftolozane/tazobactam-resistant P. aeruginosa clinical isolates. Herein, we demonstrate that the Escherichia coli strain expressing the E221K variant of PDC-3 had the highest minimum inhibitory concentrations (MICs) against a panel of ß-lactam antibiotics, including ceftolozane and ceftazidime, a cephalosporin that differs in structure largely in the R2 side chain. The kcat values of the E221K variant for both substrates were equivalent, whereas the Km for ceftolozane (341 ± 64 µM) was higher than that for ceftazidime (174 ± 20 µM). Timed mass spectrometry, thermal stability, and equilibrium unfolding studies revealed key mechanistic insights. Enhanced sampling molecular dynamics simulations identified conformational changes in the E221K variant Ω-loop, where a hidden pocket adjacent to the catalytic site opens and stabilizes ceftolozane for efficient hydrolysis. Encouragingly, the diazabicyclooctane ß-lactamase inhibitor avibactam restored susceptibility to ceftolozane and ceftazidime in cells producing the E221K variant. In addition, a boronic acid transition state inhibitor, LP-06, lowered the ceftolozane and ceftazidime MICs by 8-fold for the E221K-expressing strain. Understanding these structural changes in evolutionarily selected variants is critical toward designing effective ß-lactam/ß-lactamase inhibitor therapies for P. aeruginosa infections.IMPORTANCE The presence of ß-lactamases (e.g., PDC-3) that have naturally evolved and acquired the ability to break down ß-lactam antibiotics (e.g., ceftazidime and ceftolozane) leads to highly resistant and potentially lethal Pseudomonas aeruginosa infections. We show that wild-type PDC-3 ß-lactamase forms an acyl enzyme complex with ceftazidime, but it cannot accommodate the structurally similar ceftolozane that has a longer R2 side chain with increased basicity. A single amino acid substitution from a glutamate to a lysine at position 221 in PDC-3 (E221K) causes the tyrosine residue at 223 to adopt a new position poised for efficient hydrolysis of both cephalosporins. The importance of the mechanism of action of the E221K variant, in particular, is underscored by its evolutionary recurrences in multiple bacterial species. Understanding the biochemical and molecular basis for resistance is key to designing effective therapies and developing new ß-lactam/ß-lactamase inhibitor combinations.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cephalosporin Resistance , Cephalosporins/pharmacology , Pseudomonas aeruginosa/drug effects , Tazobactam/pharmacology , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Gene Expression , Kinetics , Mass Spectrometry , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Protein Conformation , Protein Folding , Protein Stability , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Temperature , beta-Lactamases/chemistry , beta-Lactamases/genetics
15.
ACS Infect Dis ; 4(3): 325-336, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29144724

ABSTRACT

Acinetobacter baumannii is a multidrug resistant pathogen that infects more than 12 000 patients each year in the US. Much of the resistance to ß-lactam antibiotics in Acinetobacter spp. is mediated by class C ß-lactamases known as Acinetobacter-derived cephalosporinases (ADCs). ADCs are unaffected by clinically used ß-lactam-based ß-lactamase inhibitors. In this study, five boronic acid transition state analog inhibitors (BATSIs) were evaluated for inhibition of the class C cephalosporinase ADC-7. Our goal was to explore the properties of BATSIs designed to probe the R1 binding site. Ki values ranged from low micromolar to subnanomolar, and circular dichroism (CD) demonstrated that each inhibitor stabilizes the ß-lactamase-inhibitor complexes. Additionally, X-ray crystal structures of ADC-7 in complex with five inhibitors were determined (resolutions from 1.80 to 2.09 Å). In the ADC-7/CR192 complex, the BATSI with the lowest Ki (0.45 nM) and greatest Δ Tm (+9 °C), a trifluoromethyl substituent, interacts with Arg340. Arg340 is unique to ADCs and may play an important role in the inhibition of ADC-7. The ADC-7/BATSI complexes determined in this study shed light into the unique recognition sites in ADC enzymes and also offer insight into further structure-based optimization of these inhibitors.


Subject(s)
Acinetobacter/enzymology , Boronic Acids/chemistry , Boronic Acids/pharmacology , Cephalosporinase/chemistry , Cephalosporinase/metabolism , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology , Binding Sites , Circular Dichroism , Crystallography, X-Ray , Models, Molecular , Protein Binding , Protein Conformation
16.
ACS Infect Dis ; 4(3): 337-348, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29144725

ABSTRACT

Boronic acids are attracting a lot of attention as ß-lactamase inhibitors, and in particular, compound S02030 ( Ki = 44 nM) proved to be a good lead compound against ADC-7 ( Acinetobacter-derived cephalosporinase), one of the most significant resistance determinants in A. baumannii. The atomic structure of the ADC-7/S02030 complex highlighted the importance of critical structural determinants for recognition of the boronic acids. Herein, to elucidate the role in recognition of the R2-carboxylate, which mimics the C3/C4 found in ß-lactams, we designed, synthesized, and characterized six derivatives of S02030 (3a). Out of the six compounds, the best inhibitors proved to be those with an explicit negative charge (compounds 3a-c, 3h, and 3j, Ki = 44-115 nM), which is in contrast to the derivatives where the negative charge is omitted, such as the amide derivative 3d ( Ki = 224 nM) and the hydroxyamide derivative 3e ( Ki = 155 nM). To develop a structural characterization of inhibitor binding in the active site, the X-ray crystal structures of ADC-7 in a complex with compounds 3c, SM23, and EC04 were determined. All three compounds share the same structural features as in S02030 but only differ in the carboxy-R2 side chain, thereby providing the opportunity of exploring the distinct binding mode of the negatively charged R2 side chain. This cephalosporinase demonstrates a high degree of versatility in recognition, employing different residues to directly interact with the carboxylate, thus suggesting the existence of a "carboxylate binding region" rather than a binding site in ADC enzymes. Furthermore, this class of compounds was tested against resistant clinical strains of A. baumannii and are effective at inhibiting bacterial growth in conjunction with a ß-lactam antibiotic.


Subject(s)
Acinetobacter/enzymology , Boronic Acids/chemistry , Boronic Acids/pharmacology , Cephalosporinase/chemistry , Cephalosporinase/metabolism , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology , Binding Sites , Boronic Acids/chemical synthesis , Crystallography, X-Ray , Models, Molecular , Molecular Structure , Protein Binding , Protein Conformation , beta-Lactamase Inhibitors/chemical synthesis
17.
Antimicrob Agents Chemother ; 60(3): 1760-6, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26729491

ABSTRACT

Resistance to expanded-spectrum cephalosporins and carbapenems has rendered certain strains of Klebsiella pneumoniae the most problematic pathogens infecting patients in the hospital and community. This broad-spectrum resistance to ß-lactamases emerges in part via the expression of KPC-2 and SHV-1 ß-lactamases and variants thereof. KPC-2 carbapenemase is particularly worrisome, as the genetic determinant encoding this ß-lactamase is rapidly spread via plasmids. Moreover, KPC-2, a class A enzyme, is difficult to inhibit with mechanism-based inactivators (e.g., clavulanate). In order to develop new ß-lactamase inhibitors (BLIs) to add to the limited available armamentarium that can inhibit KPC-2, we have structurally probed the boronic acid transition state analog S02030 for its inhibition of KPC-2 and SHV-1. S02030 contains a boronic acid, a thiophene, and a carboxyl triazole moiety. We present here the 1.54- and 1.87-Å resolution crystal structures of S02030 bound to SHV-1 and KPC-2 ß-lactamases, respectively, as well as a comparative analysis of the S02030 binding modes, including a previously determined S02030 class C ADC-7 ß-lactamase complex. S02030 is able to inhibit vastly different serine ß-lactamases by interacting with the conserved features of these active sites, which includes (i) forming the bond with catalytic serine via the boron atom, (ii) positioning one of the boronic acid oxygens in the oxyanion hole, and (iii) utilizing its amide moiety to make conserved interactions across the width of the active site. In addition, S02030 is able to overcome more distantly located structural differences between the ß-lactamases. This unique feature is achieved by repositioning the more polar carboxyl-triazole moiety, generated by click chemistry, to create polar interactions as well as reorient the more hydrophobic thiophene moiety. The former is aided by the unusual polar nature of the triazole ring, allowing it to potentially form a unique C-H…O 2.9-Å hydrogen bond with S130 in KPC-2.


Subject(s)
Boronic Acids/chemistry , Klebsiella pneumoniae/enzymology , Triazoles/chemistry , beta-Lactamase Inhibitors/metabolism , beta-Lactamases/chemistry , Boronic Acids/metabolism , Carbapenems/metabolism , Catalytic Domain/drug effects , Cephalosporins/metabolism , Crystallography, X-Ray , Klebsiella pneumoniae/drug effects , Protein Structure, Tertiary , Thiophenes/chemistry , Triazoles/metabolism , beta-Lactamases/metabolism
18.
Antimicrob Agents Chemother ; 60(3): 1751-9, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26729496

ABSTRACT

Boronic acid transition state inhibitors (BATSIs) are competitive, reversible ß-lactamase inhibitors (BLIs). In this study, a series of BATSIs with selectively modified regions (R1, R2, and amide group) were strategically designed and tested against representative class A ß-lactamases of Klebsiella pneumoniae, KPC-2 and SHV-1. Firstly, the R1 group of compounds 1a to 1c and 2a to 2e mimicked the side chain of cephalothin, whereas for compounds 3a to 3c, 4a, and 4b, the thiophene ring was replaced by a phenyl, typical of benzylpenicillin. Secondly, variations in the R2 groups which included substituted aryl side chains (compounds 1a, 1b, 1c, 3a, 3b, and 3c) and triazole groups (compounds 2a to 2e) were chosen to mimic the thiazolidine and dihydrothiazine ring of penicillins and cephalosporins, respectively. Thirdly, the amide backbone of the BATSI, which corresponds to the amide at C-6 or C-7 of ß-lactams, was also changed to the following bioisosteric groups: urea (compound 3b), thiourea (compound 3c), and sulfonamide (compounds 4a and 4b). Among the compounds that inhibited KPC-2 and SHV-1 ß-lactamases, nine possessed 50% inhibitory concentrations (IC50s) of ≤ 600 nM. The most active compounds contained the thiopheneacetyl group at R1 and for the chiral BATSIs, a carboxy- or hydroxy-substituted aryl group at R2. The most active sulfonamido derivative, compound 4b, lacked an R2 group. Compound 2b (S02030) was the most active, with acylation rates (k2/K) of 1.2 ± 0.2 × 10(4) M(-1) s(-1) for KPC-2 and 4.7 ± 0.6 × 10(3) M(-1) s(-1) for SHV-1, and demonstrated antimicrobial activity against Escherichia coli DH10B carrying blaSHV variants and blaKPC-2 or blaKPC-3 and against clinical strains of Klebsiella pneumoniae and E. coli producing different class A ß-lactamase genes. At most, MICs decreased from 16 to 0.5 mg/liter.


Subject(s)
Boronic Acids/pharmacology , Escherichia coli/drug effects , Klebsiella pneumoniae/drug effects , Triazoles/pharmacology , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/drug effects , Boronic Acids/chemistry , Ceftazidime/chemistry , Cephalothin/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/metabolism , Microbial Sensitivity Tests , Penicillins/chemistry , Structure-Activity Relationship
19.
European J Org Chem ; 2015(5): 1075-1083, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-26257579

ABSTRACT

Stereoselective synthesis of previously unreported 1,2,3-triazol-1-yl-methaneboronic acids has been achieved from azidomethaneboronates by Copper-catalyzed Azide-Alkyne Cycloaddition (CuAAC). The proximity of the cycloaddition reaction center to the boronic group is not detrimental for the stability of the sp3-carbon-boron bond nor to the stereoisomeric composition, further expanding the field of application of click chemistry to new boronate substrates and offering a new potential scaffold for protease inhibitors.

20.
J Med Chem ; 58(14): 5445-58, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26102369

ABSTRACT

Boronic acid transition-state inhibitors (BATSIs) represent one of the most promising classes of ß-lactamase inhibitors. Here we describe a new class of BATSIs, namely, 1-amido-2-triazolylethaneboronic acids, which were synthesized by combining the asymmetric homologation of boronates with copper-catalyzed azide-alkyne cycloaddition for the stereoselective insertion of the amido group and the regioselective formation of the 1,4-disubstituted triazole, respectively. This synthetic pathway, which avoids intermediate purifications, proved to be flexible and efficient, affording in good yields a panel of 14 BATSIs bearing three different R1 amide side chains (acetamido, benzylamido, and 2-thienylacetamido) and several R substituents on the triazole. This small library was tested against two clinically relevant class C ß-lactamases from Enterobacter spp. and Pseudomonas aeruginosa. The K(i) value of the best compound (13a) was as low as 4 nM with significant reduction of bacterial resistance to the combination of cefotaxime/13a.


Subject(s)
Boronic Acids/chemistry , Boronic Acids/pharmacology , Click Chemistry , Drug Design , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism , Escherichia coli/drug effects , Inhibitory Concentration 50 , Models, Molecular , Protein Conformation , Thermodynamics , Triazoles/chemistry , beta-Lactamases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...