Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 948
Filter
1.
Nat Commun ; 15(1): 8337, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333122

ABSTRACT

ß-lactam antibiotics have been prescribed for most bacterial infections since their discovery. However, resistance to ß-lactams, mediated by ß-lactamase (Bla) enzymes such as extended spectrum ß-lactamases (ESBLs), has become widespread. Bla inhibitors can restore the efficacy of ß-lactams against resistant bacteria, an approach which preserves existing antibiotics despite declining industry investment. However, the effects of combination treatment on selection for ß-lactam resistance are not well understood. Bla production confers both private benefits for resistant cells and public benefits which faster-growing sensitive cells can also exploit. These benefits may be differentially impacted by Bla inhibitors, leading to non-intuitive selection dynamics. In this study, we demonstrate strain-to-strain variation in effective combination doses, with complex growth dynamics in mixed populations. Using modeling, we derive a criterion for the selection outcome of combination treatment, dependent on the burden and effective private benefit of Bla production. We then use engineered strains and natural isolates to show that strong private benefits of Bla are associated with increased selection for resistance. Finally, we demonstrate that this parameter can be coarsely estimated using high-throughput phenotyping of clonal populations. Our analysis shows that quantifying the phenotypic responses of bacteria to combination treatment can facilitate resistance-minimizing optimization of treatment.


Subject(s)
Anti-Bacterial Agents , beta-Lactamases , beta-Lactamases/metabolism , beta-Lactamases/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/therapeutic use , beta-Lactam Resistance/genetics , beta-Lactam Resistance/drug effects , Microbial Sensitivity Tests , Drug Therapy, Combination , beta-Lactams/pharmacology , beta-Lactams/therapeutic use , Escherichia coli/drug effects , Escherichia coli/genetics , Humans , Selection, Genetic
2.
J Med Chem ; 67(18): 16796-16806, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39255340

ABSTRACT

While large library docking has discovered potent ligands for multiple targets, as the libraries have grown the hit lists can become dominated by rare artifacts that cheat our scoring functions. Here, we investigate rescoring top-ranked docked molecules with orthogonal methods to identify these artifacts, exploring implicit solvent models and absolute binding free energy perturbation as cross-filters. In retrospective studies, this approach deprioritized high-ranking nonbinders for nine targets while leaving true ligands relatively unaffected. We tested the method prospectively against hits from docking against AmpC ß-lactamase. We prioritized 128 high-ranking molecules for synthesis and testing, a mixture of 39 molecules flagged as likely cheaters and 89 that were plausible inhibitors. None of the predicted cheating compounds inhibited AmpC detectably, while 57% of the 89 plausible compounds did so. As our libraries continue to grow, deprioritizing docking artifacts by rescoring with orthogonal methods may find wide use.


Subject(s)
Molecular Docking Simulation , Small Molecule Libraries , beta-Lactamases , beta-Lactamases/chemistry , beta-Lactamases/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Ligands , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Artifacts , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/chemical synthesis
3.
Cells ; 13(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39195205

ABSTRACT

We investigated the activity of cefiderocol/ß-lactamase inhibitor combinations against clinical strains with different susceptibility profiles to cefiderocol to explore the potentiality of antibiotic combinations as a strategy to contain the major public health problem of multidrug-resistant (MDR) pathogens. Specifically, we evaluated the synergistic activity of cefiderocol with avibactam, sulbactam, or tazobactam on three of the most "Critical Priority" group of MDR bacteria (carbapenem-resistant Enterobacterales, Pseudomonas aeruginosa, and Acinetobacter baumannii). Clinical isolates were genomically characterized by Illumina iSeq 100. The synergy test was conducted with time-kill curve assays. Specifically, cefiderocol/avibactam, /sulbactam, or /tazobactam combinations were analyzed. Synergism was assigned if bacterial grow reduction reached 2 log10 CFU/mL. We reported the high antimicrobial activity of the cefiderocol/sulbactam combination against carbapenem-resistant Enterobacterales, P. aeruginosa, and A. baumannii; of the cefiderocol/avibactam combination against carbapenem-resistant Enterobacterales; and of the cefiderocol/tazobactam combination against carbapenem-resistant Enterobacterales and P. aeruginosa. Our results demonstrate that all ß-lactamase inhibitors (BLIs) tested are able to enhance cefiderocol antimicrobial activity, also against cefiderocol-resistant isolates. The cefiderocol/sulbactam combination emerges as the most promising combination, proving to highly enhance cefiderocol activity in all the analyzed carbapenem-resistant Gram-negative isolates, whereas the Cefiderocol/tazobactam combination resulted in being active only against carbapenem-resistant Enterobacterales and P. aeruginosa, and cefiderocol/avibactam was only active against carbapenem-resistant Enterobacterales.


Subject(s)
Anti-Bacterial Agents , Azabicyclo Compounds , Cefiderocol , Cephalosporins , Drug Synergism , Gram-Negative Bacteria , Microbial Sensitivity Tests , Sulbactam , Tazobactam , Azabicyclo Compounds/pharmacology , Tazobactam/pharmacology , Sulbactam/pharmacology , Cephalosporins/pharmacology , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Carbapenems/pharmacology , Humans , Acinetobacter baumannii/drug effects , Pseudomonas aeruginosa/drug effects , beta-Lactamase Inhibitors/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Drug Combinations
4.
Antimicrob Agents Chemother ; 68(9): e0027224, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39133022

ABSTRACT

The effectiveness of ß-lactam antibiotics is increasingly threatened by resistant bacteria that harbor hydrolytic ß-lactamase enzymes. Depending on the class of ß-lactamase present, ß-lactam hydrolysis can occur through one of two general molecular mechanisms. Metallo-ß-lactamases (MBLs) require active site Zn2+ ions, whereas serine-ß-lactamases (SBLs) deploy a catalytic serine residue. The result in both cases is drug inactivation via the opening of the ß-lactam warhead of the antibiotic. MBLs confer resistance to most ß-lactams and are non-susceptible to SBL inhibitors, including recently approved diazabicyclooctanes, such as avibactam; consequently, these enzymes represent a growing threat to public health. Aspergillomarasmine A (AMA), a fungal natural product, can rescue the activity of the ß-lactam antibiotic meropenem against MBL-expressing bacterial strains. However, the effectiveness of this ß-lactam/ß-lactamase inhibitor combination against bacteria producing multiple ß-lactamases remains unknown. We systematically investigated the efficacy of AMA/meropenem combination therapy with and without avibactam against 10 Escherichia coli and 10 Klebsiella pneumoniae laboratory strains tandemly expressing single MBL and SBL enzymes. Cell-based assays demonstrated that laboratory strains producing NDM-1 and KPC-2 carbapenemases were resistant to the AMA/meropenem combination but became drug-susceptible upon adding avibactam. We also probed these combinations against 30 clinical isolates expressing multiple ß-lactamases. E. coli, Enterobacter cloacae, and K. pneumoniae clinical isolates were more susceptible to AMA, avibactam, and meropenem than Pseudomonas aeruginosa and Acinetobacter baumannii isolates. Overall, the results demonstrate that a triple combination of AMA/avibactam/meropenem has potential for empirical treatment of infections caused by multiple ß-lactamase-producing bacteria, especially Enterobacterales.


Subject(s)
Anti-Bacterial Agents , Azabicyclo Compounds , Escherichia coli , Meropenem , Microbial Sensitivity Tests , beta-Lactamases , Azabicyclo Compounds/pharmacology , beta-Lactamases/metabolism , beta-Lactamases/genetics , Anti-Bacterial Agents/pharmacology , Meropenem/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/enzymology , beta-Lactamase Inhibitors/pharmacology , Humans , Drug Resistance, Multiple, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology , Drug Combinations , Enterobacter cloacae/drug effects , Enterobacter cloacae/enzymology , Aspartic Acid/analogs & derivatives
5.
Antimicrob Agents Chemother ; 68(9): e0075124, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39133021

ABSTRACT

Taniborbactam, a bicyclic boronate ß-lactamase inhibitor with activity against Klebsiella pneumoniae carbapenemase (KPC), Verona integron-encoded metallo-ß-lactamase (VIM), New Delhi metallo-ß-lactamase (NDM), extended-spectrum beta-lactamases (ESBLs), OXA-48, and AmpC ß-lactamases, is under clinical development in combination with cefepime. Susceptibility of 200 previously characterized carbapenem-resistant K. pneumoniae and 197 multidrug-resistant (MDR) Pseudomonas aeruginosa to cefepime-taniborbactam and comparators was determined by broth microdilution. For K. pneumoniae (192 KPC; 7 OXA-48-related), MIC90 values of ß-lactam components for cefepime-taniborbactam, ceftazidime-avibactam, and meropenem-vaborbactam were 2, 2, and 1 mg/L, respectively. For cefepime-taniborbactam, 100% and 99.5% of isolates of K. pneumoniae were inhibited at ≤16 mg/L and ≤8 mg/L, respectively, while 98.0% and 95.5% of isolates were susceptible to ceftazidime-avibactam and meropenem-vaborbactam, respectively. For P. aeruginosa, MIC90 values of ß-lactam components of cefepime-taniborbactam, ceftazidime-avibactam, ceftolozane-tazobactam, and meropenem-vaborbactam were 16, >8, >8, and >4 mg/L, respectively. Of 89 carbapenem-susceptible isolates, 100% were susceptible to ceftolozane-tazobactam, ceftazidime-avibactam, and cefepime-taniborbactam at ≤8 mg/L. Of 73 carbapenem-intermediate/resistant P. aeruginosa isolates without carbapenemases, 87.7% were susceptible to ceftolozane-tazobactam, 79.5% to ceftazidime-avibactam, and 95.9% and 83.6% to cefepime-taniborbactam at ≤16 mg/L and ≤8 mg/L, respectively. Cefepime-taniborbactam at ≤16 mg/L and ≤8 mg/L, respectively, was active against 73.3% and 46.7% of 15 VIM- and 60.0% and 35.0% of 20 KPC-producing P. aeruginosa isolates. Of all 108 carbapenem-intermediate/resistant P. aeruginosa isolates, cefepime-taniborbactam was active against 86.1% and 69.4% at ≤16 mg/L and ≤8 mg/L, respectively, compared to 59.3% for ceftolozane-tazobactam and 63.0% for ceftazidime-avibactam. Cefepime-taniborbactam had in vitro activity comparable to ceftazidime-avibactam and greater than meropenem-vaborbactam against carbapenem-resistant K. pneumoniae and carbapenem-intermediate/resistant MDR P. aeruginosa.


Subject(s)
Anti-Bacterial Agents , Cefepime , Drug Resistance, Multiple, Bacterial , Klebsiella pneumoniae , Microbial Sensitivity Tests , Pseudomonas aeruginosa , beta-Lactamase Inhibitors , Cefepime/pharmacology , Pseudomonas aeruginosa/drug effects , Klebsiella pneumoniae/drug effects , Anti-Bacterial Agents/pharmacology , beta-Lactamase Inhibitors/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Cephalosporins/pharmacology , Humans , beta-Lactamases/metabolism , beta-Lactamases/genetics , Boronic Acids/pharmacology , Carbapenems/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Ceftazidime/pharmacology , Borinic Acids/pharmacology , Drug Combinations , Azabicyclo Compounds/pharmacology , Carboxylic Acids
6.
Molecules ; 29(16)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39203022

ABSTRACT

Currently, antimicrobial resistance (AMR) is a serious health problem in the world, mainly because of the rapid spread of multidrug-resistant (MDR) bacteria. These include bacteria that produce ß-lactamases, which confer resistance to ß-lactams, the antibiotics with the most prescriptions in the world. Carbapenems are particularly noteworthy because they are considered the ultimate therapeutic option for MDR bacteria. However, this group of antibiotics can also be hydrolyzed by ß-lactamases, including metallo-ß-lactamases (MBLs), which have one or two zinc ions (Zn2+) on the active site and are resistant to common inhibitors of serine ß-lactamases, such as clavulanic acid, sulbactam, tazobactam, and avibactam. Therefore, the design of inhibitors against MBLs has been directed toward various compounds, with groups such as nitrogen, thiols, and metal-binding carboxylates, or compounds such as bicyclic boronates that mimic hydrolysis intermediates. Other compounds, such as dipicolinic acid and aspergillomarasmin A, have also been shown to inhibit MBLs by chelating Zn2+. In fact, recent inhibitors are based on Zn2+ chelation, which is an important factor in the mechanism of action of most MBL inhibitors. Therefore, in this review, we analyzed the current strategies for the design and mechanism of action of metal-ion-binding inhibitors that combat MDR bacteria.


Subject(s)
Zinc , beta-Lactamase Inhibitors , beta-Lactamases , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism , beta-Lactamases/chemistry , Zinc/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Humans , Metals/chemistry , Bacteria/drug effects , Bacteria/enzymology
7.
Microb Cell Fact ; 23(1): 221, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118086

ABSTRACT

ß-lactam resistance is a significant global public health issue. Outbreaks of bacteria resistant to extended-spectrum ß-lactams and carbapenems are serious health concerns that not only complicate medical care but also impact patient outcomes. The primary objective of this work was to express and purify two soluble recombinant representative serine ß­lactamases using Escherichia coli strain as an expression host and pET101/D as a cloning vector. Furthermore, a second objective was to evaluate the potential, innovative, and safe use of galloylquinic acid (GQA) from Copaifera lucens as a potential ß-lactamase inhibitor.In the present study, blaCTX-M-15 and blaKPC-2 represented genes encoding for serine ß-lactamases that were cloned from parent isolates of E. coli and K. pneumoniae, respectively, and expression as well as purification were performed. Moreover, susceptibility results demonstrated that recombinant cells became resistant to all test carbapenems (MICs; 64-128 µg/mL) and cephalosporins (MICs; 128-512 µg/mL). The MICs of the tested ß-lactam antibiotics were determined in combination with 4 µg/mL of GQA, clavulanic acid, or tazobactam against E. coli strains expressing CTX-M-15 or KPC-2-ß-lactamases. Interestingly, the combination with GQA resulted in an important reduction in the MIC values by 64-512-fold to the susceptible range with comparable results for other reference inhibitors. Additionally, the half-maximal inhibitory concentration of GQA was determined using nitrocefin as a ß-lactamase substrate. Data showed that the test agent was similar to tazobactam as an efficient inhibitors of the test enzymes, recording smaller IC50 values (CTX-M-15; 17.51 for tazobactam, 28.16 µg/mL for GQA however, KPC-2; 20.91 for tazobactam, 24.76 µg/mL for GQA) compared to clavulanic acid. Our work introduces GQA as a novel non-ß-lactam inhibitor, which interacts with the crucial residues involved in ß-lactam recognition and hydrolysis by non-covalent interactions, complementing the enzyme's active site. GQA markedly enhanced the potency of ß-lactams against carbapenemase and extended-spectrum ß-lactamase-producing strains, reducing the MICs of ß-lactams to the susceptible range. The ß-lactamase inhibitory activity of GQA makes it a promising lead molecule for the development of more potent ß-lactamase inhibitors.


Subject(s)
Escherichia coli , Microbial Sensitivity Tests , beta-Lactamase Inhibitors , beta-Lactamases , beta-Lactamases/metabolism , beta-Lactamases/genetics , beta-Lactamase Inhibitors/pharmacology , Escherichia coli/genetics , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/enzymology , Klebsiella pneumoniae/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/antagonists & inhibitors , Carbapenems/pharmacology
8.
J Antimicrob Chemother ; 79(10): 2591-2597, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39073766

ABSTRACT

OBJECTIVES: We aimed to compare the stability of the newly developed ß-lactams (cefiderocol) and ß-lactam/ß-lactamase inhibitor combinations (ceftazidime/avibactam, ceftolozane/tazobactam, aztreonam/avibactam, cefepime/taniborbactam, cefepime/zidebactam, imipenem/relebactam, meropenem/vaborbactam, meropenem/nacubactam and meropenem/xeruborbactam) against the most clinically relevant mechanisms of mutational and transferable ß-lactam resistance in Pseudomonas aeruginosa. METHODS: We screened a collection of 61 P. aeruginosa PAO1 derivatives. Eighteen isolates displayed the most relevant mechanisms of mutational resistance to ß-lactams. The other 43 constructs expressed transferable ß-lactamases from genes cloned in pUCP-24. MICs were determined by reference broth microdilution. RESULTS: Cefiderocol and imipenem/relebactam exhibited excellent in vitro activity against all of the mutational resistance mechanisms studied. Aztreonam/avibactam, cefepime/taniborbactam, cefepime/zidebactam, meropenem/vaborbactam, meropenem/nacubactam and meropenem/xeruborbactam proved to be more vulnerable to mutational events, especially to overexpression of efflux operons. The agents exhibiting the widest spectrum of activity against transferable ß-lactamases were aztreonam/avibactam and cefepime/zidebactam, followed by cefepime/taniborbactam, cefiderocol, meropenem/xeruborbactam and meropenem/nacubactam. However, some MBLs, particularly NDM enzymes, may affect their activity. Combined production of certain enzymes (e.g. NDM-1) with increased MexAB-OprM-mediated efflux and OprD deficiency results in resistance to almost all agents tested, including last options such as aztreonam/avibactam and cefiderocol. CONCLUSIONS: Cefiderocol and new ß-lactam/ß-lactamase inhibitor combinations show promising and complementary in vitro activity against mutational and transferable P. aeruginosa ß-lactam resistance. However, the combined effects of efflux pumps, OprD deficiency and efficient ß-lactamases could still result in the loss of all therapeutic options. Resistance surveillance, judicious use of new agents and continued drug development efforts are encouraged.


Subject(s)
Anti-Bacterial Agents , Azabicyclo Compounds , Cefiderocol , Cephalosporins , Drug Combinations , Microbial Sensitivity Tests , Pseudomonas aeruginosa , beta-Lactamase Inhibitors , beta-Lactamases , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/enzymology , Cephalosporins/pharmacology , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/genetics , beta-Lactamases/metabolism , Anti-Bacterial Agents/pharmacology , Azabicyclo Compounds/pharmacology , Cyclooctanes/pharmacology , Tazobactam/pharmacology , beta-Lactams/pharmacology , Humans , beta-Lactam Resistance/genetics , Ceftazidime/pharmacology , Pseudomonas Infections/microbiology , Pseudomonas Infections/drug therapy , Gene Transfer, Horizontal , Chromosomes, Bacterial/genetics
9.
Eur J Clin Microbiol Infect Dis ; 43(9): 1787-1794, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38995343

ABSTRACT

PURPOSE: Carbapenem resistant Pseudomonas aeruginosa (CR-PA) is escalating worldwide and leaves clinicians few therapeutic options in recent years, ß-lactam/ß-lactamase inhibitor combinations (ceftolozane-tazobactam, ceftazidime-avibactam) and a new siderophore cephalosporin (cefiderocol) have been approved for the treatment of P. aeruginosa infection and have shown potent activity against isolates defined as carbapenem resistant. The aim of this study was to determine the phenotypic profile of these agents against CR-PA in the emerging setting of carbapenemases. METHODS: CR-PA clinical isolates were collected from three teaching hospitals in different geographical regions between January 2017-December 2021. All isolates were subjected to phenotypic carbapenemase testing using modified carbapenem inactivation method. MICs were determined by reference broth microdilution and evaluated according to EUCAST standards, while genotypic profiling was determined using PCR methods. RESULTS: 244 CR-PA sourced most frequently from the respiratory tract (32.2%), blood (20.4%) and urine (17.5%) were evaluated. Of all isolates, 32 (13.1%) were phenotypically and 38 (15.6%) were genotypically defined as carbapenemase-positive. The most common carbapenemase was GES (63.1%), followed by VIM (15.8%). The MIC50/90(S%) of ceftazidime/avibactam, ceftolozane/tazobactam and cefiderocol in all CR-PA isolates were 4 and 32 (80%), 1 and > 64 (69%) and 0.25 and 1 mg/L (96%), respectively. Cefiderocol was also the most active agent in carbapenemase-positive isolates (90%). CONSLUSION: While ceftolozane/tazobactam and ceftazidime/avibactam remained highly active against CR-PA devoid of carbapenemases, cefiderocol provided potent in vitro activity irrespective of carbapenemase production. When considering the potential clinical utility of newer agents against CR-PA, regional variations in carbapenemase prevalence must be considered.


Subject(s)
Anti-Bacterial Agents , Azabicyclo Compounds , Cefiderocol , Ceftazidime , Cephalosporins , Drug Combinations , Microbial Sensitivity Tests , Pseudomonas Infections , Pseudomonas aeruginosa , Tazobactam , Humans , Cephalosporins/pharmacology , Ceftazidime/pharmacology , Azabicyclo Compounds/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/isolation & purification , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/enzymology , Pseudomonas Infections/microbiology , Tazobactam/pharmacology , Anti-Bacterial Agents/pharmacology , Middle Aged , Female , Male , Adult , beta-Lactamases/genetics , beta-Lactamases/metabolism , Aged , Carbapenems/pharmacology , Bacterial Proteins/genetics , Young Adult , Adolescent , beta-Lactamase Inhibitors/pharmacology , Child
10.
Pharmacotherapy ; 44(8): 658-674, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38949413

ABSTRACT

Antibiotic resistance has become a global threat as it is continuously growing due to the evolution of ß-lactamases diminishing the activity of classic ß-lactam (BL) antibiotics. Recent antibiotic discovery and development efforts have led to the availability of ß-lactamase inhibitors (BLIs) with activity against extended-spectrum ß-lactamases as well as Klebsiella pneumoniae carbapenemase (KPC)-producing carbapenem-resistant organisms (CRO). Nevertheless, there is still a lack of drugs that target metallo-ß-lactamases (MBL), which hydrolyze carbapenems efficiently, and oxacillinases (OXA) often present in carbapenem-resistant Acinetobacter baumannii. This review aims to provide a snapshot of microbiology, pharmacology, and clinical data for currently available BL/BLI treatment options as well as agents in late stage development for CRO harboring various ß-lactamases including MBL and OXA-enzymes.


Subject(s)
Anti-Bacterial Agents , Carbapenems , Gram-Negative Bacteria , beta-Lactamase Inhibitors , beta-Lactams , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/therapeutic use , beta-Lactams/pharmacology , beta-Lactams/therapeutic use , Carbapenems/pharmacology , Carbapenems/therapeutic use , Gram-Negative Bacteria/drug effects , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , beta-Lactam Resistance , beta-Lactamases
11.
Antimicrob Agents Chemother ; 68(8): e0172123, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38990013

ABSTRACT

The use of ß-lactam/ß-lactamase inhibitors constitutes an important strategy to counteract ß-lactamases in multidrug-resistant (MDR) Gram-negative bacteria. Recent reports have described ceftazidime-/avibactam-resistant isolates producing CTX-M variants with different amino acid substitutions (e.g., P167S, L169Q, and S130G). Relebactam (REL) combined with imipenem has proved very effective against Enterobacterales producing ESBLs, serine-carbapenemases, and AmpCs. Herein, we evaluated the inhibitory efficacy of REL against CTX-M-96, a CTX-M-15-type variant. The CTX-M-96 structure was obtained in complex with REL at 1.03 Å resolution (PDB 8EHH). REL was covalently bound to the S70-Oγ atom upon cleavage of the C7-N6 bond. Compared with apo CTX-M-96, binding of REL forces a slight displacement of the deacylating water inwards the active site (0.81 Å), making the E166 and N170 side chains shift to create a proper hydrogen bonding network. Binding of REL also disturbs the hydrophobic patch formed by Y105, P107, and Y129, likely due to the piperidine ring of REL that creates clashes with these residues. Also, a remarkable change in the positioning of the N104 sidechain is also affected by the piperidine ring. Therefore, differences in the kinetic behavior of REL against class A ß-lactamases seem to rely, at least in part, on differences in the residues being involved in the association and stabilization of the inhibitor before hydrolysis. Our data provide the biochemical and structural basis for REL effectiveness against CTX-M-producing Gram-negative pathogens and essential details for further DBO design. Imipenem/REL remains an important choice for dealing with isolates co-producing CTX-M with other ß-lactamases.


Subject(s)
Azabicyclo Compounds , beta-Lactamase Inhibitors , beta-Lactamases , Azabicyclo Compounds/pharmacology , Azabicyclo Compounds/chemistry , beta-Lactamases/genetics , beta-Lactamases/metabolism , beta-Lactamases/chemistry , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/chemistry , Crystallography, X-Ray , Anti-Bacterial Agents/pharmacology , Imipenem/pharmacology , Imipenem/chemistry , Ceftazidime/pharmacology , Microbial Sensitivity Tests , Catalytic Domain
12.
Int J Antimicrob Agents ; 64(3): 107268, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38972552

ABSTRACT

The global public health threat of antibiotic resistance continues to escalate, and necessitates the implementation of urgent measures to expand the arsenal of antimicrobial drugs. This study identified a benzoxaborane compound, namely 5-chloro-1,3-dihydro-1-hydroxy-2,1-benzoxaborole (AN2178), which can inhibit the catalytic activity of the Klebsiella pneumoniae carbapenemase (KPC-2) enzyme effectively. The efficacy of AN2718 as an inhibitor for the KPC-2 enzyme was verified through various assays, including enzyme activity assays and isothermal titration calorimetry. Results of multiple biochemical assays, minimum inhibitory concentration assays and time-killing assays also showed that binding of AN2718 to KPC-2 enabled restoration of the bactericidal effect of meropenem. The survival rate of mice infected with carbapenem-resistant, high-virulence strains increased significantly upon treatment with AN2718. Most importantly, the meropenem and AN2718 combination was effective on KPC-2 mutations such as KPC-33, which evolved clinically and exhibited resistance to ceftazidime-avibactam after clinical use for a couple of years. Comprehensive safety tests both in vitro and in vivo, such as cytotoxicity, haemolytic activity and cytochrome P450 inhibition assays, demonstrated that AN2718 was safe for clinical use. These promising data indicate that AN2718 has high potential for approval for the treatment of drug resistant-bacterial infections, including those caused by ceftazidime-avibactam-resistant strains. AN2718 can be regarded as a valuable addition to the current antimicrobial armamentarium, and a promising tool to combat antimicrobial resistance.


Subject(s)
Anti-Bacterial Agents , Carbapenem-Resistant Enterobacteriaceae , Klebsiella Infections , Klebsiella pneumoniae , Meropenem , Animals , Female , Humans , Mice , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/genetics , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/therapeutic use , beta-Lactamases/genetics , beta-Lactamases/metabolism , Carbapenem-Resistant Enterobacteriaceae/drug effects , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/genetics , Meropenem/pharmacology , Meropenem/therapeutic use , Microbial Sensitivity Tests
13.
Nature ; 632(8023): 39-49, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085542

ABSTRACT

In this Review, we explore natural product antibiotics that do more than simply inhibit an active site of an essential enzyme. We review these compounds to provide inspiration for the design of much-needed new antibacterial agents, and examine the complex mechanisms that have evolved to effectively target bacteria, including covalent binders, inhibitors of resistance, compounds that utilize self-promoted entry, those that evade resistance, prodrugs, target corrupters, inhibitors of 'undruggable' targets, compounds that form supramolecular complexes, and selective membrane-acting agents. These are exemplified by ß-lactams that bind covalently to inhibit transpeptidases and ß-lactamases, siderophore chimeras that hijack import mechanisms to smuggle antibiotics into the cell, compounds that are activated by bacterial enzymes to produce reactive molecules, and antibiotics such as aminoglycosides that corrupt, rather than merely inhibit, their targets. Some of these mechanisms are highly sophisticated, such as the preformed ß-strands of darobactins that target the undruggable ß-barrel chaperone BamA, or teixobactin, which binds to a precursor of peptidoglycan and then forms a supramolecular structure that damages the membrane, impeding the emergence of resistance. Many of the compounds exhibit more than one notable feature, such as resistance evasion and target corruption. Understanding the surprising complexity of the best antimicrobial compounds provides a roadmap for developing novel compounds to address the antimicrobial resistance crisis by mining for new natural products and inspiring us to design similarly sophisticated antibiotics.


Subject(s)
Anti-Bacterial Agents , Bacteria , Biological Products , Animals , Humans , Aminoglycosides/pharmacology , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bacteria/drug effects , Bacteria/enzymology , Bacteria/metabolism , beta Lactam Antibiotics/chemistry , beta Lactam Antibiotics/pharmacology , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology , Biological Products/chemistry , Biological Products/pharmacology , Biological Products/metabolism , Drug Design , Drug Resistance, Bacterial/drug effects , Peptidyl Transferases/antagonists & inhibitors , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/metabolism , Siderophores/metabolism , Siderophores/chemistry , Siderophores/pharmacology
14.
Int J Antimicrob Agents ; 64(3): 107266, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38971203

ABSTRACT

BACKGROUND: Novel beta-lactams show activity against many multidrug-resistant Gram-negative bacteria that cause severe lung infections. Understanding pharmacokinetic/pharmacodynamic characteristics of these agents may help optimise outcomes in the treatment of pneumonia. OBJECTIVES: To describe and appraise studies that report pulmonary pharmacokinetic and pharmacodynamic data of cefiderocol, ceftazidime/avibactam, ceftolozane/tazobactam, imipenem/cilastatin/relebactam and meropenem/vaborbactam. METHODS: MEDLINE (PubMed), Embase, Web of Science and Scopus libraries were used for the literature search. Pulmonary population pharmacokinetic and pharmacokinetic/pharmacodynamic studies on adult patients receiving cefiderocol, ceftazidime/avibactam, ceftolozane/tazobactam, imipenem/cilastatin/relebactam, and meropenem/vaborbactam published in peer-reviewed journals were included. Two independent authors screened, reviewed and extracted data from included articles. A reporting guideline for clinical pharmacokinetic studies (ClinPK statement) was used for bias assessment. Relevant outcomes were included, such as population pharmacokinetic parameters and probability of target attainment of dosing regimens. RESULTS: Twenty-four articles were included. There was heterogeneity in study methods and reporting of results, with diversity across studies in adhering to the ClinPK statement checklist. Ceftolozane/tazobactam was the most studied agent. Only two studies collected epithelial lining fluid samples from patients with pneumonia. All the other phase I studies enrolled healthy subjects. Significant population heterogeneity was evident among available population pharmacokinetic models. Probabilities of target attainment rates above 90% using current licensed dosing regiments were reported in most studies. CONCLUSIONS: Although lung pharmacokinetics was rarely described, this review observed high target attainment using plasma pharmacokinetic data for all novel beta-lactams. Future studies should describe lung pharmacokinetics in patient populations at risk of carbapenem-resistant pathogen infections.


Subject(s)
Anti-Bacterial Agents , Cephalosporins , Drug Combinations , Gram-Negative Bacteria , beta-Lactamase Inhibitors , beta-Lactams , Humans , beta-Lactamase Inhibitors/pharmacokinetics , beta-Lactamase Inhibitors/therapeutic use , beta-Lactamase Inhibitors/pharmacology , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , beta-Lactams/pharmacokinetics , beta-Lactams/therapeutic use , beta-Lactams/pharmacology , Cephalosporins/pharmacokinetics , Cephalosporins/therapeutic use , Cephalosporins/pharmacology , Gram-Negative Bacteria/drug effects , Tazobactam/pharmacokinetics , Tazobactam/therapeutic use , Tazobactam/pharmacology , Pneumonia, Bacterial/drug therapy , Azabicyclo Compounds/pharmacokinetics , Azabicyclo Compounds/therapeutic use , Azabicyclo Compounds/pharmacology , Carbapenems/pharmacokinetics , Carbapenems/therapeutic use , Carbapenems/pharmacology , Gram-Negative Bacterial Infections/drug therapy , Ceftazidime/pharmacokinetics , Ceftazidime/therapeutic use , Cefiderocol , Meropenem/pharmacokinetics , Meropenem/therapeutic use , Meropenem/pharmacology , Imipenem/pharmacokinetics , Imipenem/therapeutic use , Drug Resistance, Multiple, Bacterial , Microbial Sensitivity Tests , Cilastatin, Imipenem Drug Combination/pharmacokinetics , Cilastatin, Imipenem Drug Combination/therapeutic use , Boronic Acids , Heterocyclic Compounds, 1-Ring
15.
Microbiol Spectr ; 12(8): e0426623, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38934607

ABSTRACT

New ß-lactam-ß-lactamase inhibitor combinations represent last-resort antibiotics to treat infections caused by multidrug-resistant Pseudomonas aeruginosa. Carbapenemase gene acquisition can limit their spectrum of activity, and reports of resistance toward these new molecules are increasing. In this multi-center study, we evaluated the prevalence of resistance to ceftazidime-avibactam (CZA) and comparators among P. aeruginosa clinical isolates from bloodstream infections, hospital-acquired or ventilator-associated pneumonia, and urinary tract infections, circulating in Southern Italy. We also investigated the clonality and content of relevant ß-lactam resistance mechanisms of CZA-resistant (CZAR) isolates. A total of 120 P. aeruginosa isolates were collected. CZA was among the most active ß-lactams, retaining susceptibility in the 81.7% of cases, preceded by cefiderocol (95.8%) and followed by ceftolozane-tazobactam (79.2%), meropenem-vaborbactam (76.1%), imipenem-relebactam (75%), and aztreonam (69.6%). Among non-ß-lactams, colistin and amikacin were active against 100% and 85.8% of isolates respectively. In CZAR strains subjected to whole-genome sequencing (n = 18), resistance was mainly due to the expression of metallo-ß-lactamases (66.6% VIM-type and 5.5% FIM-1), followed by PER-1 (16.6%) and GES-1 (5.5%) extended-spectrum ß-lactamases, mostly carried by international high-risk clones (ST111 and ST235). Of note, two strains producing the PER-1 enzyme were resistant to all ß-lactams, including cefiderocol. In conclusion, the CZA resistance rate among P. aeruginosa clinical isolates in Southern Italy remained low. CZAR isolates were mostly metallo-ß-lactamases producers and belonging to ST111 and ST253 epidemic clones. It is important to implement robust surveillance systems to monitor emergence of new resistance mechanisms and to limit the spread of P. aeruginosa high-risk clones. IMPORTANCE: Multidrug-resistant Pseudomonas aeruginosa infections are a growing threat due to the limited therapeutic options available. Ceftazidime-avibactam (CZA) is among the last-resort antibiotics for the treatment of difficult-to-treat P. aeruginosa infections, although resistance due to the acquisition of transferable ß-lactamase genes is increasing. With this work, we report that CZA represents a highly active antipseudomonal ß-lactam compound (after cefiderocol), and that metallo-ß-lactamases (VIM-type) and extended-spectrum ß-lactamases (GES and PER-type) production is the major factor underlying CZA resistance in isolates from Southern Italian hospitals. In addition, we reported that such resistance mechanisms were mainly carried by the international high-risk clones ST111 and ST235.


Subject(s)
Anti-Bacterial Agents , Azabicyclo Compounds , Ceftazidime , Drug Combinations , Drug Resistance, Multiple, Bacterial , Microbial Sensitivity Tests , Pseudomonas Infections , Pseudomonas aeruginosa , Azabicyclo Compounds/pharmacology , Ceftazidime/pharmacology , Humans , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/isolation & purification , Anti-Bacterial Agents/pharmacology , Pseudomonas Infections/microbiology , Pseudomonas Infections/drug therapy , Pseudomonas Infections/epidemiology , Drug Resistance, Multiple, Bacterial/genetics , Italy/epidemiology , beta-Lactamase Inhibitors/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Male , Female , beta-Lactamases/genetics , beta-Lactamases/metabolism , Middle Aged , beta-Lactams/pharmacology , Aged , Adult
17.
J Glob Antimicrob Resist ; 38: 216-222, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38908824

ABSTRACT

OBJECTIVES: Ceftolozane-tazobactam (C/T) is a combination of a cephalosporin and a ß-lactamase inhibitor with activity against Gram-negative bacilli (GNB). The study aims were to evaluate the activity of C/T in vitro vs. comparators against clinical GNB isolated from Chinese paediatric patients. METHODS: From 2017-2021, 660 GNB isolates were collected from 20 hospitals across China. The minimum inhibitory concentrations were tested using a Trek Diagnostic System (Thermo Fisher Scientific). Susceptibility was determined by CLSI broth microdilution and the results were interpreted according to CLSI M100 (2021) breakpoints. RESULTS: GNB isolates were obtained from paediatric patients < 18 years old, mainly from the bloodstream (n = 146), intraperitoneal cavity (n = 138), lower respiratory (n = 278) and urinary tract (n = 96). Overall, C/T was active against 76.6% of 436 Enterobacterales, with a descending susceptibility rate of 100.0% to S. marcescens, 92.2% to E. coli, 83.3% to K. oxytoca, 66.7% to K. aerogenes, 66.7% to P. mirabilis, 58.6% to K. pneumoniae and 57.1% to E. cloacae. The susceptibility of P. aeruginosa to C/T was 89.4%, which was the highest among the ß-lactam antibiotics and was second only to amikacin (92.9%). Isolates of respiratory tract infection (RTI) derived P. aeruginosa were highly susceptible (93.8%) to C/T, while <75% of isolates of RTI derived P. aeruginosa were susceptible to the other ß-lactam antibiotics tested, except for ceftazidime-avibactam (91.2%). CONCLUSION: GNBs collected from paediatric patients in China showed a high susceptibility to C/T making this drug combination an effective choice for treating the paediatric population, especially those infected with P. aeruginosa.


Subject(s)
Anti-Bacterial Agents , Cephalosporins , Gram-Negative Bacteria , Gram-Negative Bacterial Infections , Microbial Sensitivity Tests , Tazobactam , Humans , Child , Cephalosporins/pharmacology , Tazobactam/pharmacology , China/epidemiology , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/isolation & purification , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/epidemiology , Child, Preschool , Anti-Bacterial Agents/pharmacology , Infant , Adolescent , Female , Male , beta-Lactamase Inhibitors/pharmacology , Infant, Newborn
18.
J Glob Antimicrob Resist ; 38: 247-251, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38936472

ABSTRACT

INTRODUCTION: Novel beta-lactam/beta-lactamase inhibitor (BIBLI) combinations are commercially available and have been used for treating carbapenem-resistant Klebsiella pneumoniae (CRKP) infections. Continuous surveillance of susceptibility profiles and resistance mechanism identification are necessary to monitor the evolution of resistance within these agents. OBJECTIVE: The purpose of this study was to evaluate the susceptibility rates of ceftazidime/avibactam, imipenem/relebactam and meropenem/vaborbactam in CRKP isolated from patients with bloodstream infections who underwent screening for a randomized clinical trial in Brazil. METHODS: Minimum inhibitory concentrations (MICs) were determined for meropenem, ceftazidime/avibactam, imipenem/relebactam and meropenem/vaborbactam using the gradient diffusion strip method. Carbapenemase genes were detected by multiplex real-time polymerase chain reaction. Klebsiella pneumoniae carbapenemase (KPC)-producing isolates showing resistance to any BLBLI and New Delhi Metallo-beta-lactamase (NDM)-producing isolates with susceptibility to any BLBLI isolates were further submitted for whole-genome sequencing. RESULTS: From a total of 69 CRKP isolates, 39 were positive for blaKPC, 19 for blaNDM and 11 for blaKPC and blaNDM. KPC-producing isolates demonstrated susceptibility rates above 94 % for all BLBLIs. Two isolates with resistance to meropenem/vaborbactam demonstrated a Gly and Asp duplication at the porin OmpK36 as well as a truncated OmpK35. All NDM-producing isolates, including KPC and NDM coproducers, demonstrated susceptibility rates to ceftazidime/avibactam, imipenem/relebactam and meropenem/vaborbactam of 0 %, 9.1-21.1 % and 9.1-26.3 %, respectively. Five NDM-producing isolates that presented susceptibility to BLBLIs also had porin alterations CONCLUSIONS: This study showed that, although high susceptibility rates to BLBLIs were found, KPC-2 isolates were able to demonstrate resistance probably as a result of porin mutations. Additionally, NDM-1 isolates showed susceptibility to BLBLIs in vitro.


Subject(s)
Anti-Bacterial Agents , Azabicyclo Compounds , Carbapenem-Resistant Enterobacteriaceae , Ceftazidime , Drug Combinations , Klebsiella Infections , Klebsiella pneumoniae , Microbial Sensitivity Tests , beta-Lactamase Inhibitors , beta-Lactamases , Humans , Brazil , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/isolation & purification , beta-Lactamase Inhibitors/pharmacology , Klebsiella Infections/microbiology , Azabicyclo Compounds/pharmacology , Ceftazidime/pharmacology , beta-Lactamases/genetics , Anti-Bacterial Agents/pharmacology , Carbapenem-Resistant Enterobacteriaceae/drug effects , Carbapenem-Resistant Enterobacteriaceae/genetics , Carbapenem-Resistant Enterobacteriaceae/isolation & purification , Bacterial Proteins/genetics , Meropenem/pharmacology , Imipenem/pharmacology , Bacteremia/microbiology , Boronic Acids/pharmacology , Heterocyclic Compounds, 1-Ring
20.
J Antimicrob Chemother ; 79(8): 1914-1918, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38943535

ABSTRACT

OBJECTIVES: Mycobacterium abscessus has emerged as an opportunistic pathogen responsible for lung infections, especially in cystic fibrosis patients. In spite of the production of the broad-spectrum ß-lactamase BlaMab, the carbapenem imipenem is recommended in the initial phase of the treatment of pulmonary infections. Here, we determine whether the addition of vaborbactam, a second-generation ß-lactamase inhibitor belonging to the boronate family, improves the activity of ß-lactams against M. abscessus. METHODS: The activity of ß-lactams, alone or in combination with vaborbactam, was evaluated against M. abscessus CIP104536 by determining MICs, time-killing and intramacrophage activity. Kinetic parameters for the inhibition of BlaMab by vaborbactam were determined by spectrophotometry. RESULTS: The combination of vaborbactam (8 mg/L) with ß-lactams decreased more than 8 times the MIC of amoxicillin (from >1024 to 128 mg/L) and 2 times the MICs of meropenem (from 16 to 8 mg/L) and imipenem (from 4 to 2 mg/L). The reduction of the MICs was less than that obtained with avibactam at 4 mg/L for amoxicillin (from >1024 to 16 mg/L, more than 64 times less) and for meropenem (from 16 to 4 mg/L, 4 times less). In vitro and intracellularly, M. abscessus was not killed by the meropenem/vaborbactam combination, in spite of significant in vitro inhibition of BlaMab by vaborbactam. CONCLUSIONS: Inhibition of BlaMab by vaborbactam decreases the MIC of ß-lactams, including that of meropenem. As meropenem/vaborbactam is clinically available, this combination offers an alternative therapeutic option that should be evaluated for the treatment of pulmonary infections due to M. abscessus.


Subject(s)
Anti-Bacterial Agents , Boronic Acids , Microbial Sensitivity Tests , Mycobacterium abscessus , beta-Lactams , Mycobacterium abscessus/drug effects , Anti-Bacterial Agents/pharmacology , beta-Lactams/pharmacology , Boronic Acids/pharmacology , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium Infections, Nontuberculous/drug therapy , Drug Synergism , beta-Lactamase Inhibitors/pharmacology , Humans , Animals , Mice , Microbial Viability/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL