Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.296
Filter
1.
Nat Commun ; 15(1): 3947, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38729951

ABSTRACT

Gram-negative bacteria (GNB) are a major cause of neonatal sepsis in low- and middle-income countries (LMICs). Although the World Health Organization (WHO) reports that over 80% of these sepsis deaths could be prevented through improved treatment, the efficacy of the currently recommended first- and second-line treatment regimens for this condition is increasingly affected by high rates of drug resistance. Here we assess three well known antibiotics, fosfomycin, flomoxef and amikacin, in combination as potential antibiotic treatment regimens by investigating the drug resistance and genetic profiles of commonly isolated GNB causing neonatal sepsis in LMICs. The five most prevalent bacterial isolates in the NeoOBS study (NCT03721302) are Klebsiella pneumoniae, Acinetobacter baumannii, E. coli, Serratia marcescens and Enterobacter cloacae complex. Among these isolates, high levels of ESBL and carbapenemase encoding genes are detected along with resistance to ampicillin, gentamicin and cefotaxime, the current WHO recommended empiric regimens. The three new combinations show excellent in vitro activity against ESBL-producing K. pneumoniae and E. coli isolates. Our data should further inform and support the clinical evaluation of these three antibiotic combinations for the treatment of neonatal sepsis in areas with high rates of multidrug-resistant Gram-negative bacteria.


Subject(s)
Acinetobacter baumannii , Anti-Bacterial Agents , Gram-Negative Bacteria , Gram-Negative Bacterial Infections , Klebsiella pneumoniae , Microbial Sensitivity Tests , Neonatal Sepsis , Humans , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Neonatal Sepsis/microbiology , Neonatal Sepsis/drug therapy , Infant, Newborn , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/isolation & purification , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/isolation & purification , Acinetobacter baumannii/genetics , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/isolation & purification , Klebsiella pneumoniae/genetics , Amikacin/pharmacology , Amikacin/therapeutic use , Fosfomycin/pharmacology , Fosfomycin/therapeutic use , beta-Lactamases/genetics , beta-Lactamases/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/isolation & purification , Developing Countries , Drug Resistance, Multiple, Bacterial/genetics , Drug Therapy, Combination , Serratia marcescens/drug effects , Serratia marcescens/genetics , Serratia marcescens/isolation & purification , Enterobacter cloacae/drug effects , Enterobacter cloacae/genetics , Enterobacter cloacae/isolation & purification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
2.
ACS Infect Dis ; 10(5): 1711-1724, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38562022

ABSTRACT

Fosfomycin is a broad-spectrum single-dose therapy approved for treating lower urinary tract infections. Acinetobacter baumannii, one of the five major UTI-causing pathogens, is intrinsically resistant to fosfomycin. Reduced uptake and active efflux are major reasons for this intrinsic resistance. AbaF, a major facilitator superfamily class of transporter in A. baumannii, is responsible for fosfomycin efflux and biofilm formation. This study describes the identification and validation of a novel small-molecule efflux pump inhibitor that potentiates fosfomycin efficacy against A. baumannii. An AbaF inhibitor screening was performed against Escherichia coli KAM32/pUC18_abaF, using the noninhibitory concentration of 24 putative efflux pump inhibitors. The inhibitory activity of IITR08367 [bis(4-methylbenzyl) disufide] against fosfomycin/H+ antiport was validated using ethidium bromide efflux, quinacrine-based proton-sensitive fluorescence, and membrane depolarization assays. IITR08367 inhibits fosfomycin/H+ antiport activity by perturbing the transmembrane proton gradient. IITR08367 is a nontoxic molecule that potentiates fosfomycin activity against clinical strains of A. baumannii and prevents biofilm formation by inhibiting efflux pump (AbaF). The IITR08367-fosfomycin combination reduced bacterial burden by > 3 log10 in kidney and bladder tissue in the murine UTI model. Overall, fosfomycin, in combination with IITR08367, holds the potential to treat urinary tract infections caused by A. baumannii.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Anti-Bacterial Agents , Biofilms , Drug Synergism , Fosfomycin , Microbial Sensitivity Tests , Acinetobacter baumannii/drug effects , Fosfomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Animals , Mice , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Biofilms/drug effects , Membrane Transport Proteins/metabolism , Female , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology
3.
Mikrobiyol Bul ; 58(2): 113-124, 2024 Apr.
Article in Turkish | MEDLINE | ID: mdl-38676580

ABSTRACT

The aim of this study was to investigate the detection of teicoplanin and fosfomycin antibiotic susceptibility of methicillin-resistant Staphylococcus aureus (MRSA) strains by different methods and to evaluate the antibacterial synergistic effect of teicoplanin-fosfomycin combination by using checkerboard assay and time kill curve assay. Forty-five MRSA strains isolated from clinical samples in routine medical microbiology laboratory of Göztepe Prof. Dr. Süleyman Yalçin City Hospital were included in the study. In the first stage of the combination study, minimum inhibitory concentrations (MIC) were investigated by broth microdilution for teicoplanin and by both broth microdilution and agar dilution methods for fosfomycin. The combination of teicoplanin and fosfomycin was tested by the checkerboard method in 45 MRSA strains and combination effect was determined according to fractional inhibitory concentration index (ΣFIC) calculation. The synergistic effect and bactericidal activity of antibiotic combination were studied against a randomly selected strain from the strains used in the study by using time-kill method for 24 hours. As a result of teicoplanin and fosfomycin antibiotic susceptibility studies, all isolates were found to be susceptible to both antibiotics according to the susceptibility breakpoints determined by the European Committee on Antimicrobial Susceptibility Testing (EUCAST). A synergistic effect was found in 22 (49%), additive effect in 22 (49%) and indifferent effect in one (2%) of the 45 strains studied with the checkerboard method. The mean ΣFIC of 45 isolates was found to be 0.5. In the combination study of the antibiotics of the isolate that was studied with time-kill method, synergism was detected for 1/8 MIC concentrations at 12th hour and 24th hour and synergism at 1/4 MIC concentration at sixth hour, 12th hour and 24th hour. In the combination study of 1/4 MIC concentrations of antibiotics, bactericidal effect was detected at sixth hour and this effect was observed to disappear at 12th and 24th hours. High rate of synergistic antibacterial effect of teicoplanin-fosfomycin combination on MRSA isolates was demonstrated as a result of in vitro tests. Such studies conducted on antibiotic-resistant bacterial infections will provide clinicians different treatment options and will contribute to increasing survival. As a result of this study, provided that it is supported by future clinical studies, it can be stated that the teicoplanin-fosfomycin combination may be an effective treatment option in community and hospital-acquired infections caused by MRSA.


Subject(s)
Anti-Bacterial Agents , Drug Synergism , Fosfomycin , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Staphylococcal Infections , Teicoplanin , Fosfomycin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Teicoplanin/pharmacology , Anti-Bacterial Agents/pharmacology , Humans , Staphylococcal Infections/microbiology
4.
Int J Mol Sci ; 25(8)2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38674148

ABSTRACT

It is now generally accepted that the success of antitumor therapy can be impaired by concurrent antibiotic therapy, the presence of certain bacteria, and elevated defensin levels around the tumor tissue. The aim of our current investigation was to identify the underlying changes in microbiome and defensin levels in the tumor tissue induced by different antibiotics, as well as the duration of this modification. The microbiome of the tumor tissues was significantly different from that of healthy volunteers. Comparing only the tumor samples, no significant difference was confirmed between the untreated group and the group treated with antibiotics more than 3 months earlier. However, antibiotic treatment within 3 months of analysis resulted in a significantly modified microbiome composition. Irrespective of whether Fosfomycin, Fluoroquinolone or Beta-lactam treatment was used, the abundance of Bacteroides decreased, and Staphylococcus abundance increased. Large amounts of the genus Acinetobacter were observed in the Fluoroquinolone-treated group. Regardless of the antibiotic treatment, hBD1 expression of the tumor cells consistently doubled. The increase in hBD2 and hBD3 expression was the highest in the Beta-lactam treated group. Apparently, antibiotic treatment within 3 months of sample analysis induced microbiome changes and defensin expression levels, depending on the identity of the applied antibiotic.


Subject(s)
Anti-Bacterial Agents , Microbiota , Urinary Bladder Neoplasms , beta-Defensins , Humans , beta-Defensins/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/microbiology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Microbiota/drug effects , Male , Female , Middle Aged , Aged , Fosfomycin/therapeutic use , Fosfomycin/pharmacology , Fluoroquinolones/therapeutic use , Fluoroquinolones/pharmacology , beta-Lactams/therapeutic use , beta-Lactams/pharmacology
5.
ACS Infect Dis ; 10(5): 1739-1752, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38647213

ABSTRACT

Reverse analogs of the phosphonohydroxamic acid antibiotic fosmidomycin are potent inhibitors of the nonmevalonate isoprenoid biosynthesis enzyme 1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR, IspC) of Plasmodium falciparum. Some novel analogs with large phenylalkyl substituents at the hydroxamic acid nitrogen exhibit nanomolar PfDXR inhibition and potent in vitro growth inhibition of P. falciparum parasites coupled with good parasite selectivity. X-ray crystallographic studies demonstrated that the N-phenylpropyl substituent of the newly developed lead compound 13e is accommodated in a subpocket within the DXR catalytic domain but does not reach the NADPH binding pocket of the N-terminal domain. As shown for reverse carba and thia analogs, PfDXR selectively binds the S-enantiomer of the new lead compound. In addition, some representatives of the novel inhibitor subclass are nanomolar Escherichia coli DXR inhibitors, whereas the inhibition of Mycobacterium tuberculosis DXR is considerably weaker.


Subject(s)
Aldose-Ketose Isomerases , Antimalarials , Fosfomycin , Hydroxamic Acids , Multienzyme Complexes , Plasmodium falciparum , Fosfomycin/pharmacology , Fosfomycin/analogs & derivatives , Fosfomycin/chemistry , Aldose-Ketose Isomerases/antagonists & inhibitors , Aldose-Ketose Isomerases/metabolism , Aldose-Ketose Isomerases/chemistry , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Hydroxamic Acids/pharmacology , Hydroxamic Acids/chemistry , Antimalarials/pharmacology , Antimalarials/chemistry , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Multienzyme Complexes/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Structure-Activity Relationship , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/enzymology , Models, Molecular , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Catalytic Domain , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism
6.
BMC Microbiol ; 24(1): 95, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38519885

ABSTRACT

BACKGROUND: The emergence of antimicrobial resistance in bacterial pathogens is a growing concern worldwide due to its impact on the treatment of bacterial infections. The "Trojan Horse" strategy has been proposed as a potential solution to overcome drug resistance caused by permeability issues. OBJECTIVE: The objective of our research was to investigate the bactericidal activity and mechanism of action of the "Trojan Horse" strategy using enterobactin conjugated with Ciprofloxacin and Fosfomycin against the antibiotic-resistant Escherichia coli strain OQ866153. METHODOLOGY: Enterobactin, a mixed ligand of E. coli OQ866153, was conjugated with Ciprofloxacin and Fosfomycin individually to aid active absorption via specific enterobactin binding proteins (FepABCDG). The effectiveness of the conjugates was assessed by measuring their bactericidal activity against E. coli OQ866153, as well as their ability to inhibit DNA gyrase enzyme and biofilm formation. RESULTS: The Fe+3-enterobactin-Ciprofloxacin conjugate effectively inhibited the DNA gyrase enzyme (Docking score = -8.597 kcal/mol) and resulted in a lower concentration (25 µg/ml) required to eliminate supercoiled DNA plasmids compared to the parent drug (35 µg/ml; Docking score = -6.264 kcal/mol). The Fe+3-Enterobactin-Fosfomycin conjugate showed a higher inhibition percentage (100%) of biofilm formation compared to Fosfomycin (21.58%) at a concentration of 2 mg/ml, with docking scores of -5.481 and -3.756 kcal/mol against UDP-N acetylglucosamine 1-carboxyvinyltransferase MurA. CONCLUSION: The findings of this study suggest that the "Trojan Horse" strategy using enterobactin conjugated with Ciprofloxacin and Fosfomycin can effectively overcome permeability issues caused by efflux proteins and enhance the bactericidal activity of these drugs against antibiotic-resistant strains of E. coli.


Subject(s)
Anti-Bacterial Agents , Fosfomycin , Anti-Bacterial Agents/chemistry , Fosfomycin/pharmacology , Ciprofloxacin/pharmacology , Escherichia coli , Enterobactin/chemistry , Enterobactin/metabolism , Enterobactin/pharmacology , DNA Gyrase , Microbial Sensitivity Tests
8.
J Agric Food Chem ; 72(14): 7684-7693, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38532701

ABSTRACT

Fosmidomycin (FOS) is a natural product inhibiting the DXR enzyme in the MEP pathway and has stimulated interest for finding more suitable FOS analogues. Herein, two series of FOS analogue hydroxamate-containing bisphosphonates as proherbicides were designed, with bisphosphonate replacing the phosphonic unit in FOS while retaining the hydroxamate (BPF series) or replacing it with retro-hydroxamate (BPRF series). The BPF series were synthesized through a three-step reaction sequence including Michael addition of vinylidenebisphosphonate, N-acylation, and deprotection, and the BPRF series were synthesized with a retro-Claisen condensation incorporated into the reaction sequence. Evaluation on model plants demonstrated several compounds having considerable herbicidal activities, and in particular, compound 8m exhibited multifold activity enhancement as compared to the control FOS. The proherbicide properties were comparatively validated. Furthermore, DXR enzyme assay, dimethylallyl pyrophosphate rescue, and molecular docking verified 8m to be a promising proherbicide candidate targeting the DXR enzyme. In addition, 8m also displayed good antimalarial activities.


Subject(s)
Aldose-Ketose Isomerases , Antimalarials , Fosfomycin , Fosfomycin/analogs & derivatives , Diphosphonates , Molecular Docking Simulation , Fosfomycin/pharmacology , Aldose-Ketose Isomerases/metabolism
9.
Urogynecology (Phila) ; 30(3): 205-213, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38484233

ABSTRACT

IMPORTANCE: Urinary tract infections (UTIs) are common in older-aged women. Our study examined bacterial persistence with commonly prescribed antibiotics. Bacterial growth was demonstrated despite antibiotic treatment. OBJECTIVES: The aims of this study were to quantify the bacterial persister phenotype in urine collected from postmenopausal women with acute and recurrent UTI and to determine the capabilities of first-line antibiotics to effectively treat persister cells. STUDY DESIGN: This was an institutional review board-approved cross-sectional analysis within a large academic referral center. Uropathogens were cultured from postmenopausal women with acute or recurrent UTI and screened for persister cells using persistence assays. Demographic and clinical variables were collected and analyzed. The entire experimental process was repeated in triplicate. Data were analyzed for significance (P < 0.05) between the persister culture and antibiotic treatments using a 1-way analysis of variance with multiple comparisons in Prism 9.3.0. RESULTS: Forty participants were included: 62.5% White, 22.5% Black, 3% Asian, and 2% Hispanic with a mean age of 72.3 ± 11.62 years. The persister phenotype was demonstrated in all of Escherichia coli isolates. Treatment with fosfomycin demonstrated reduced colony-forming units per milliliter compared with control (P < 0.01). Among recurrent isolates, there was a statistically significant decrease in colony-forming units per milliliter after antibiotic treatment with all 4 antibiotics (P < 0.05). CONCLUSIONS: This study demonstrated in vitro bacterial persistence in uropathogens from urogynecology patients despite treatment with commonly prescribed antibiotics. Fosfomycin generated the least amount of persister cells. Results suggest that persistence may be one bacterial defense mechanism involved in UTIs. Further research is needed to understand the clinical implications.


Subject(s)
Fosfomycin , Urinary Tract Infections , Humans , Female , Middle Aged , Aged , Aged, 80 and over , Fosfomycin/pharmacology , Cross-Sectional Studies , Postmenopause , Urinary Tract Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Escherichia coli/genetics
10.
Int J Antimicrob Agents ; 63(5): 107120, 2024 May.
Article in English | MEDLINE | ID: mdl-38417705

ABSTRACT

OBJECTIVES: This study aimed to appraise clinical practice guidelines (CPGs) for the treatment of carbapenem-resistant Gram-negative Bacilli (CRGNB) infections and to summarise the recommendations. METHODS: A systematic search of the literature published from January 2012 to March 2023 was undertaken to identify CPGs related to CRGNB infections treatment. The methodological and reporting quality of eligible CPGs were assessed using six domains of the Appraisal of Guidelines for Research and Evaluation (AGREE) II tool and seven domains of the Reporting Items for practice Guidelines in HealThcare (RIGHT) checklist. Basic information and recommendations of included CPGs were extracted and compared. RESULTS: A total of 21 CPGs from 7953 relevant articles were included. The mean overall AGREE II score was 62.7%, and was highest for "clarity of presentation" (90.2%) and lowest for "stakeholder involvement" (44.8%). The overall reporting quality of all of the CPGs was suboptimal, with the proportion of eligible items ranging from 45.7 to 85.7%. The treatment of CRGNB infections is related to the type of pathogen, the sensitivity of antimicrobial agents, and the site of infection. In general, the recommended options mainly included novel ß-lactam/ ß-lactamase inhibitors, cefiderocol, ampicillin-sulbactam (mainly for carbapenem-resistant Acinetobacter baumannii [CRAB]), and combination therapy, involving polymyxin B/colistin, tigecycline (except for carbapenem-resistant Pseudomonas aeruginosa), aminoglycosides, carbapenems, fosfomycin, and sulbactam (mainly for CRAB). CONCLUSIONS: The methodological and reporting quality of CPGs for the treatment of CRGNB infections are generally suboptimal and need further improvement. Both monotherapy with novel drugs and combination therapy play important roles in the treatment.


Subject(s)
Anti-Bacterial Agents , Carbapenems , Gram-Negative Bacteria , Gram-Negative Bacterial Infections , Practice Guidelines as Topic , Humans , Carbapenems/therapeutic use , Carbapenems/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacteria/drug effects , Acinetobacter baumannii/drug effects , Tigecycline/therapeutic use , Tigecycline/pharmacology , Sulbactam/therapeutic use , Sulbactam/pharmacology , Microbial Sensitivity Tests/standards , Cefiderocol , Fosfomycin/therapeutic use , Fosfomycin/pharmacology
11.
J Vet Med Sci ; 86(4): 413-420, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38346727

ABSTRACT

Fosfomycin (FOM) is an approved veterinary medicinal product for large animals in Japan, but Clinical breakpoint (CBP) for antimicrobial susceptibility test (AST) is not defined for animals. This study aimed at conducting a pharmacokinetics/pharmacodynamics (PK/PD) analysis to determine the PK/PD cutoff for the CBP in horses. Drug concentrations following single intravenous administration (IV) of 20 mg/kg body weight (BW) FOM in nine horses were measured using liquid chromatography/mass spectrometry. The data were modelled using a nonlinear mixed-effects model, followed by Monte Carlo simulations. A 90% probability of target attainment for a PK/PD target of the ratio of Area Under the free plasma concentration-time curve divided by the minimal inhibitory concentration (MIC) >24 hr was set as PK/PD cut-off. The PK/PD cutoff for FOM 20 mg/kg BW q12 hr IV was estimated with the MIC value of ≤16.0 mg/L, and this regimen was considered effective against E. coli (MIC90; 16.0 mg/L) in healthy horses based on the MIC90 values of the wild population. Owing to the relevance of FOM to human health, veterinarians should use q 12 hr FOM 20 mg /kg against E. coli infections with an MIC <16 µg/mL, as suggested by our PK/PD cutoff after AST.


Subject(s)
Escherichia coli Infections , Fosfomycin , Horse Diseases , Humans , Animals , Horses , Fosfomycin/pharmacology , Fosfomycin/therapeutic use , Anti-Bacterial Agents/therapeutic use , Escherichia coli , Monte Carlo Method , Escherichia coli Infections/veterinary , Microbial Sensitivity Tests/veterinary , Horse Diseases/drug therapy
12.
Article in English | MEDLINE | ID: mdl-38324871

ABSTRACT

Extended-spectrum beta-lactamase producing and ciprofloxacin-non-susceptible Escherichia coli are clinical and environmental issues. We evaluated the susceptibility profile of fosfomycin in non-susceptible E. coli isolated from urine and the environment. We measured the activity of fosfomycin against 319 and 36 E. coli strains from urine and environmental isolates, respectively, collected from rivers. Fosfomycin resistance profiles were investigated using the minimal inhibitory concentration (MIC), according to the Clinical and Laboratory Standards Institute (CLSI) and the European Committee for Antimicrobial Susceptibility Testing (EUCAST) guidelines. Antibiotic susceptibility testing revealed that 5% and 6.6% of urine samples were non-susceptible to fosfomycin according to CLSI and EUCAST guidelines, respectively. The fosfomycin MIC50/90 was 0.5/4 mg/L. Of the 36 E. coli isolates from river water, 11.1% and 13,8% were non-susceptible to fosfomycin according to CLSI and EUCAST, respectively (range ≤0.25 ≥512 mg/L). All the isolates with MIC ≥512 mg/L for fosfomycin showed the fosA3 gene. Fosfomycin resistance was more frequent in the environment than in clinical samples.


Subject(s)
Escherichia coli Infections , Fosfomycin , Humans , Fosfomycin/pharmacology , Ciprofloxacin/pharmacology , Escherichia coli/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Escherichia coli Infections/drug therapy , beta-Lactamases/genetics , Microbial Sensitivity Tests
13.
Antimicrob Agents Chemother ; 68(1): e0080323, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38078906

ABSTRACT

IMPORTANCE: While fosfomycin resistance is rare, the observation of non-susceptible subpopulations among clinical Escherichia coli isolates is a common phenomenon during antimicrobial susceptibility testing (AST) in American and European clinical labs. Previous evidence suggests that mutations eliciting this phenotype are of high biological cost to the pathogen during infection, leading to current recommendations of neglecting non-susceptible colonies during AST. Here, we report that the most common route to fosfomycin resistance, as well as novel routes described in this work, does not impair virulence in uropathogenic E. coli, the major cause of urinary tract infections, suggesting a re-evaluation of current susceptibility guidelines is warranted.


Subject(s)
Escherichia coli Infections , Fosfomycin , Urinary Tract Infections , Uropathogenic Escherichia coli , Humans , Fosfomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/genetics
14.
Microbiol Spectr ; 12(1): e0331823, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38063387

ABSTRACT

IMPORTANCE: Mechanistic understanding of pharmacodynamic interactions is key for the development of rational antibiotic combination therapies to increase efficacy and suppress the development of resistances. Potent tools to provide those insights into pharmacodynamic drug interactions are semi-mechanistic modeling and simulation techniques. This study uses those techniques to provide a detailed understanding with regard to the direction and strength of the synergy of ceftazidime-avibactam and ceftazidime-fosfomycin in a clinical Escherichia coli isolate expressing extended spectrum beta-lactamase (CTX-M-15 and TEM-4) and carbapenemase (OXA-244) genes. Enhanced killing effects in combination were identified as a driver of the synergy and were translated from static time-kill experiments into the dynamic hollow fiber infection model. These findings in combination with a suppression of the emergence of resistance in combination emphasize a potential clinical benefit with regard to increased efficacy or to allow for dose reductions with maintained effect sizes to avoid toxicity.


Subject(s)
Azabicyclo Compounds , Ceftazidime , Fosfomycin , Ceftazidime/pharmacology , Ceftazidime/therapeutic use , Fosfomycin/pharmacology , Escherichia coli/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , beta-Lactamases/genetics , Microbial Sensitivity Tests , Drug Combinations
16.
Eur J Clin Microbiol Infect Dis ; 43(1): 17-31, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37975976

ABSTRACT

PURPOSE: Vancomycin-resistant enterococci (VRE) are a leading cause of hospital-acquired infections with limited therapeutic options. Combination of at least two antimicrobials is a possible strategy to obtain rapid and sustained bactericidal effects and overcome the emergence of resistance. We revised the literature on linezolid synergistic properties from in vitro studies to assess its activity in combination with molecules belonging to other antibiotic classes against Enterococcus spp. METHODS: We performed a systematic review of the literature from three peer-reviewed databases including papers evaluating linezolid synergistic properties in vitro against Enterococcus spp. isolates. RESULTS: We included 206 Enterococcus spp. isolates (92 E. faecalis, 90 E. faecium, 2 E. gallinarum, 3 E. casseliflavus, 19 Enterococcus spp.) from 24 studies. When an isolate was tested with different combinations, each combination was considered independently for further analysis. The most frequent interaction was indifferent effect (247/343, 72% of total interactions). The highest synergism rates were observed when linezolid was tested in combination with rifampin (10/49, 20.4% of interactions) and fosfomycin (16/84, 19.0%, of interactions). Antagonistic effect accounted for 7/343 (2.0%) of total interactions. CONCLUSION: Our study reported overall limited synergistic in vitro properties of linezolid with other antibiotics when tested against Enterococcus spp. The clinical choice of linezolid in combination with other antibiotics should be guided by reasoned empiric therapy in the suspicion of a polymicrobial infection or targeted therapy on microbiological results, rather than on an intended synergistic effect of the linezolid-based combination.


Subject(s)
Enterococcus faecium , Fosfomycin , Gram-Positive Bacterial Infections , Vancomycin-Resistant Enterococci , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Enterococcus faecalis , Fosfomycin/pharmacology , Fosfomycin/therapeutic use , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/microbiology , Linezolid/pharmacology , Linezolid/therapeutic use , Microbial Sensitivity Tests , Rifampin/pharmacology , Rifampin/therapeutic use
18.
Pest Manag Sci ; 80(2): 846-856, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37794283

ABSTRACT

BACKGROUND: The enzymes involved in the 2-C-methyl-d-erythritol 4-phosphate (MEP) pathway are attractive targets of a new mode of action for developing anti-infective drugs and herbicides, and inhibitors against 1-deoxy-d-xylulose 5-phosphate reductoisomerase (IspC), the second key enzyme in the pathway, have been intensively investigated; however, few works are reported regarding IspC inhibitors designed for new herbicide discovery. RESULTS: A series of fosmidomycin (FOS) analogs were designed with nitrogen-containing linkers replacing the trimethylene linker between the two active substructures of FOS, phosphonic acid and hydroxamic acid. Synthesis followed a facile three-step route of sequential aza-Michael addition of α-amino acids to dibenzyl vinylphosphonate, amidation of the amino acid carboxyl with O-benzyl hydroxylamine, and simultaneous removal of the benzyl protective groups. Biological activity evaluation of IspC and model plants revealed that some compounds had moderate enzyme and model plant growth inhibition effects. In particular, compound 10g, which has a N-(4-fluorophenylethyl) nitrogen-containing linker, exhibited the best plant inhibition activities, superior to the control FOS against the model plants Arabidopsis thaliana, Brassica napus L., Amaranthus retroflexus and Echinochloa crus-galli. A dimethylallyl pyrophosphate rescue assay on A. thaliana confirmed that both 10g and FOS exert their herbicidal activity by blocking the MEP pathway. This result consistent with molecular docking, which confirmed 10g and FOS binding to the IspC active site in a similar way. CONCLUSION: Compound 10g has excellent herbicidal activity and represents the first herbicide lead structure of a new mode of action that targets IspC enzyme in the MEP pathway. © 2023 Society of Chemical Industry.


Subject(s)
Erythritol/analogs & derivatives , Fosfomycin , Herbicides , Sugar Phosphates , Molecular Docking Simulation , Fosfomycin/pharmacology , Herbicides/chemistry , Nitrogen
19.
Int J Antimicrob Agents ; 63(1): 107027, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37926273

ABSTRACT

The inducible inner membrane transporters, UhpT and GlpT are considered to be unique fosfomycin transporters. Glucose-6-phosphate, the substrate for UhpT, enhances fosfomycin activity. Previous work indicates that the fructose phosphotransferase system (PTS) might be involved in fosfomycin transport in the bacterial species, Stenotrophomonas maltophilia. Fosfomycin transport in Escherichia coli has been extensively studied and characterised. The current paper addresses the potential fosfomycin transport activity of the fructose PTS in E. coli. Notably, the deletion of both fructose-specific and general PTS proteins in E. coli increases fosfomycin resistance, which indicates that fructose PTS is involved in fosfomycin transport in E. coli. Further, although inactivation of UhpT, the canonical fosfomycin transporter, in E. coli increases fosfomycin resistance by 2-fold, inactivation of genes encoding the PTS increases it by up to 256-fold. Moreover, intracellular accumulation declines in the absence of both transporters, being mutations in the PTS associated with a larger decline. The results presented in this paper re-open the study of fosfomycin transport and reveal the role of the PTS in the transport of this bactericidal antibiotic in E. coli.


Subject(s)
Escherichia coli Proteins , Fosfomycin , Fosfomycin/pharmacology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Protein Kinases/genetics , Fructose/metabolism
20.
ACS Infect Dis ; 10(1): 127-137, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38104323

ABSTRACT

The antibiotic fosfomycin (FOS) is widely recognized for the treatment of lower urinary tract infections with Escherichia coli and has lately gained importance as a therapeutic option to combat multidrug-resistant bacteria. However, resistance to FOS frequently develops through mutations reducing its uptake. Although the inner-membrane transport of FOS has been extensively studied in E. coli, its outer-membrane (OM) transport remains insufficiently understood. While evaluating minimal inhibitory concentrations in OM porin-deficient mutants, we observed that the E. coli ΔompFΔompC strain is four times more resistant to FOS than the wild type and the respective single mutants. Continuous monitoring of FOS-induced lysis of porin-deficient strains additionally highlighted the importance of LamB. The relevance of OmpF, OmpC, and LamB to FOS uptake was confirmed by electrophysiological and transcriptional analysis. Our study gives for the first time in-depth insight into the transport of FOS through the OM in E. coli.


Subject(s)
Escherichia coli Proteins , Fosfomycin , Escherichia coli/genetics , Escherichia coli/metabolism , Fosfomycin/pharmacology , Biological Transport , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Porins/genetics , Porins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...