Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
1.
J Antimicrob Chemother ; 79(6): 1372-1379, 2024 06 03.
Article in English | MEDLINE | ID: mdl-38597137

ABSTRACT

BACKGROUND: IV fosfomycin is used against MDR Gram-negative bacilli (GNB) but has dose-limiting side effects, especially in patients with impaired kidney function. OBJECTIVES: To determine the optimal dosage of IV fosfomycin for patients with varying degrees of kidney function. METHODS: Adult patients receiving IV fosfomycin for treatment of GNB were eligible. Five serial blood samples were collected after at least three doses of fosfomycin; plasma was assayed by LC-MS/MS and modelled by population pharmacokinetic analysis. The PTA for AUC24/MIC of 98.9 for Escherichia coli and Klebsiella pneumoniae, and 40.8 for Pseudomonas aeruginosa were computed by Monte Carlo simulations. Cumulative fractions of response (CFR) were analysed for each pathogen using EUCAST MIC distributions. RESULTS: A total of 24 patients were included. Creatinine clearance (CLCR) and gender significantly influenced fosfomycin clearance. The kidney function-adjusted dosing regimens are proposed by using the lowest dose that can achieve ≥90% PTA for AUC24/MIC of 98.9 at an MIC of ≤32 mg/L (EUCAST v.13 susceptibility breakpoint for Enterobacterales). For patients with normal kidney function (CLCR 91-120 mL/min), a dosage of 15 g/day is suggested. This regimen achieved 97.1% CFR against E. coli, whereas CFR was 72.9% for K. pneumoniae and 76.7% for P. aeruginosa. CONCLUSIONS: A fosfomycin dosage of 15 g/day with adjustment according to kidney function provided high PTA and CFR when treating E. coli. This dosage is lower than that used in current practice and may improve tolerability. Higher dosages may be needed for P. aeruginosa; however, safety data are limited.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Multiple, Bacterial , Fosfomycin , Gram-Negative Bacterial Infections , Klebsiella pneumoniae , Microbial Sensitivity Tests , Pseudomonas aeruginosa , Humans , Fosfomycin/pharmacokinetics , Fosfomycin/administration & dosage , Fosfomycin/pharmacology , Fosfomycin/adverse effects , Female , Male , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Middle Aged , Aged , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Adult , Klebsiella pneumoniae/drug effects , Pseudomonas aeruginosa/drug effects , Escherichia coli/drug effects , Aged, 80 and over , Administration, Intravenous , Monte Carlo Method , Tandem Mass Spectrometry , Gram-Negative Bacteria/drug effects
2.
CPT Pharmacometrics Syst Pharmacol ; 13(3): 464-475, 2024 03.
Article in English | MEDLINE | ID: mdl-38108548

ABSTRACT

Antimicrobial resistance increasingly complicates neonatal sepsis in a global context. Fosfomycin and amikacin are two agents being tested in an ongoing multicenter neonatal sepsis trial. Although neonatal pharmacokinetics (PKs) have been described for these drugs, the physiological variability within neonatal populations makes population PKs in this group uncertain. Physiologically-based pharmacokinetic (PBPK) models were developed in Simcyp for fosfomycin and amikacin sequentially for adult, pediatric, and neonatal populations, with visual and quantitative validation compared to observed data at each stage. Simulations were performed using the final validated neonatal models to determine drug exposures for each drug across a demographic range, with probability of target attainment (PTA) assessments. Successfully validated neonatal PBPK models were developed for both fosfomycin and amikacin. PTA analysis demonstrated high probability of target attainment for amikacin 15 mg/kg i.v. q24h and fosfomycin 100 mg/kg (in neonates aged 0-7 days) or 150 mg/kg (in neonates aged 7-28 days) i.v. q12h for Enterobacterales with fosfomycin and amikacin minimum inhibitory concentrations at the adult breakpoints. Repeat analysis in premature populations demonstrated the same result. PTA analysis for a proposed combination fosfomycin-amikacin target was also performed. The simulated regimens, tested in a neonatal sepsis trial, are likely to be adequate for neonates across different postnatal ages and gestational age. This work demonstrates a template for determining target attainment for antimicrobials (alone or in combination) in special populations without sufficient available PK data to otherwise assess with traditional pharmacometric methods.


Subject(s)
Fosfomycin , Neonatal Sepsis , Humans , Infant, Newborn , Amikacin/pharmacokinetics , Anti-Bacterial Agents/pharmacokinetics , Fosfomycin/pharmacokinetics , Microbial Sensitivity Tests , Neonatal Sepsis/drug therapy , Multicenter Studies as Topic , Clinical Trials as Topic
3.
Microbiol Spectr ; 9(3): e0087121, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34851157

ABSTRACT

The rapid spread of antibiotic resistance among Enterococcus has prompted considerable interest in determining the dosage regimen of linezolid combined with fosfomycin. A checkerboard assay was employed to evaluate whether linezolid combined with fosfomycin had a synergistic effect on Enterococcus isolates from the hospital, including three drug-resistant strains (MIC of linezolid [MICLZD], ≥8 mg/L; MIC of fosfomycin [MICFOF], ≥256 mg/L). The in vitro static time-kill assay, dynamic pharmacokinetic (PK)/pharmacodynamic (PD) model, and semimechanistic PK/PD model were used to explore and predict effective combined dosage regimens. The checkerboard assay and in vitro static time-kill assay demonstrated that linezolid combined with fosfomycin has a synergistic effect on drug-resistant and sensitive Enterococcus. In the in vitro PK/PD model, the dosage regimen of linezolid (8 mg/L or 12 mg/L, steady-state concentration) combined with fosfomycin (6 g or 8 g) via a 0.5-h infusion every 8 h effectively suppressed bacterial growth at 24 h with a 3 log10 CFU/mL decrease compared with the initial inocula against two resistant and one sensitive Enterococcus isolates. The semimechanistic PK/PD model predicted that linezolid (more than 16 mg/L) combined with fosfomycin (6 g or 10 g) via a 0.5-h infusion every 8 h was required to achieve a 4 log10 CFU/mL decrease at 24 h against Enterococcus isolates (MICLZD ≥ 8 mg/L and MICFOF ≥ 256 mg/L). According to the prediction of the semimechanical PK/PD model, the effect of the combination was driven by linezolid, with fosfomycin enhancing the effect. Our study is the first to explore the synergistic effects of these two drugs from a qualitative and quantitative perspective and provides a simulation tool for future studies. IMPORTANCE In this study, we found that linezolid combined with fosfomycin could kill Enterococcus in vitro and that the administered dose was significantly lower after the combination treatment, which could reduce adverse effects and the development of drug resistance. The potential mechanism of the two-drug combination against Enterococcus was revealed from a quantitative perspective, which is an important step toward dose optimization in simulated humans. We hope that our research will help build a better relationship between clinicians and patients as we work together to address the challenges of antibiotic resistance in the 21st century.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/pharmacokinetics , Enterococcus/drug effects , Fosfomycin/pharmacology , Gram-Positive Bacterial Infections/drug therapy , Linezolid/pharmacology , Cross Infection/drug therapy , Cross Infection/microbiology , Drug Resistance, Bacterial/genetics , Drug Synergism , Enterococcus/genetics , Enterococcus/growth & development , Fosfomycin/pharmacokinetics , Gram-Positive Bacterial Infections/microbiology , Humans , Linezolid/pharmacokinetics , Microbial Sensitivity Tests
4.
J Antimicrob Chemother ; 77(1): 169-173, 2021 12 24.
Article in English | MEDLINE | ID: mdl-34586373

ABSTRACT

BACKGROUND: Fosfomycin is used increasingly in the treatment of MDR bacteria. It is eliminated by renal excretion, but data regarding dosing recommendations for patients undergoing modern means of renal replacement therapies are scarce. OBJECTIVES: Evaluation of the pharmacokinetics (PK) of fosfomycin in patients undergoing prolonged intermittent renal replacement therapy (PIRRT) to guide dosing recommendations. METHODS: Fosfomycin was given in 11 (7 female) patients with severe infections undergoing PIRRT. Plasma levels were measured at several timepoints on the first day of fosfomycin therapy, as well as 5-6 days into therapy, before and after the dialyser, to calculate its clearance. Fosfomycin was measured in the collected spent dialysate. RESULTS: The median (IQR) plasma dialyser clearance for fosfomycin was 183.4 (156.9-214.9) mL/min, eliminating a total amount of 8834 (4556-10 440) mg of fosfomycin, i.e. 73.9% (45.3%-93.5%) of the initial dose. During PIRRT, the fosfomycin half-life was 2.5 (2.2-3.4) h. Data from multiple-dose PK showed an increase in fosfomycin Cmax from 266.8 (166.3-438.1) to 926.1 (446.8-1168.0) mg/L and AUC0-14 from 2540.5 (1815.2-3644.3) to 6714 (4060.6-10612.6) mg·h/L. Dialysis intensity during the study was 1.5 L/h. T>MIC was 100% in all patients. CONCLUSIONS: Patients undergoing PIRRT experience significant fosfomycin elimination, requiring a dose of 5 g/8 h to reach adequate plasma levels. However, drug accumulation may occur, depending on dialysis frequency and intensity.


Subject(s)
Fosfomycin , Intermittent Renal Replacement Therapy , Anti-Bacterial Agents/therapeutic use , Female , Fosfomycin/pharmacokinetics , Humans , Renal Dialysis , Renal Replacement Therapy
5.
BMC Infect Dis ; 21(1): 307, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33771113

ABSTRACT

BACKGROUND: Limited clinical studies describe the pharmacodynamics of fosfomycin (FOS), tigecycline (TGC) and colistin methanesulfonate (CMS) in combination against KPC-producing Klebsiella pneumoniae (KPC-Kp). Population pharmacokinetic models were used in our study. Monte Carlo simulation was conducted to calculate probability of target attainment (PTA) and cumulative fraction of response (CFR) of each agent alone and in combination against KPC-Kp in patients with normal or decreased renal function. RESULTS: The simulated regimen of FOS 6 g q8h reached ≥90% PTA against a MIC of 64 mg/L in patients with normal renal function. For patients with renal impairment, FOS 4 g q8h could provide sufficient antimicrobial coverage against a MIC of 128 mg/L. And increasing the daily dose could result to the cut-off value to 256 mg/L in decreased renal function. For TGC, conventional dosing regimens failed to reach 90% PTA against a MIC of 2 mg/L. Higher loading and daily doses (TGC 200/400 mg loading doses followed by 100 mg q12h/200 mg q24h) were needed. For CMS, none achieved 90% PTA against a MIC of 2 mg/L in normal renal function. Against KPC-Kp, the regimens of 200/400 mg loading dose followed by 100 q12h /200 mg q24h achieved > 80% CFRs regardless of renal function, followed by CMS 9 million IU loading dose followed by 4.5/3 million IU q12h in combination with FOS 8 g q8h (CFR 75-91%). CONCLUSIONS: The use of a loading dose and high daily dose of TGC and CMS in combination with FOS can provide sufficient antimicrobial coverage against critically ill patients infected with KPC-Kp.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Kidney/physiopathology , Klebsiella Infections/drug therapy , Aged , Aged, 80 and over , Anti-Bacterial Agents/therapeutic use , Carbapenem-Resistant Enterobacteriaceae/drug effects , Colistin/pharmacokinetics , Colistin/therapeutic use , Critical Illness , Female , Fosfomycin/pharmacokinetics , Fosfomycin/therapeutic use , Humans , Kidney Function Tests , Klebsiella pneumoniae/drug effects , Male , Microbial Sensitivity Tests , Middle Aged , Monte Carlo Method , Tigecycline/pharmacokinetics , Tigecycline/therapeutic use
6.
Eur J Clin Microbiol Infect Dis ; 40(6): 1117-1126, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33604721

ABSTRACT

Fosfomycin (FOS) administered intravenously has been recently rediscovered for the treatment of systemic infections due to multidrug-resistant bacteria. Its pharmacokinetic properties suggest a time-dependent dosing schedule with more clinical benefits from prolonged (PI) or continuous infusion (CI) than from intermittent infusion. We revised literature concerning PI and CI FOS to identify the best dosing regimen based on current evidence. We performed a MEDLINE/PubMed search. Ninety-one studies and their pertinent references were screened. Seventeen studies were included in the present review. The activity of FOS against Gram-negative and Gram-positive bacteria was evaluated in fourteen and five studies, respectively. Six studies evaluated FOS activity in combination with another antibiotic. Daily dosing of 12, 16, 18 or 24 g, administered with different schedules, were investigated. These regimens resulted active against the tested isolates in most cases. Emergence of resistant isolates has been shown to be preventable through the coadministration of another active antibiotic. FOS is a promising option to treat systemic infections caused by multidrug-resistant bacteria. Coadministration with another active molecule is required to prevent the emergence of resistant bacterial strains. The results of our review suggest that a therapeutic regimen including a loading dose of FOS 8 g followed by a daily dose of 16 g or 24 g CI could be the best therapeutic approach for patients with normal renal function. The dosing regimens in patients with renal insufficiency and CI or PI superiority compared with intermittent infusion in clinical settings should be further investigated.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Bacterial Infections/drug therapy , Fosfomycin/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Bacteria/drug effects , Bacteria/growth & development , Bacterial Infections/microbiology , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Multiple, Bacterial , Fosfomycin/pharmacokinetics , Humans , Infusions, Intravenous , Time Factors
7.
Microb Drug Resist ; 27(4): 546-552, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32898467

ABSTRACT

Background and Objective: Combination therapy may be a treatment option against carbapenem-resistant Acinetobacter baumannii (CR-AB) infections. In this study, we explored the utility of fosfomycin in combination with meropenem (FOS/MEM) against CR-AB isolates. Materials and Methods: Screening of synergistic activity of FOS/MEM was performed using the checkerboard assay. A pharmacokinetic/pharmacodynamic analysis was performed for various FOS/MEM regimens using Monte Carlo simulations. Results: The minimum inhibitory concentration (MIC) required to inhibit the growth of 50% of the isolates (MIC50) and MIC required to inhibit the growth of 90% of the isolates (MIC90) of FOS and MEM were reduced fourfold and twofold, respectively. The combination was synergistic against 14/50 isolates. No antagonism was observed. Sixteen out of fifty isolates had MEM MICs of ≤8 mg/L when subjected to combination therapy, compared to none with monotherapy. Forty-one out of 50 isolates had FOS MICs of ≤128 mg/L when subjected to combination therapy, compared to 17/50 isolates with monotherapy. The cumulative fraction response for MEM and FOS improved from 0% to 40% and 40% to 80%, with combination therapy, respectively. Conclusions: Addition of MEM improved the in vitro activity of FOS against the CR-AB isolates. FOS/MEM could be a plausible option to treat CR-AB for a small fraction of isolates.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Fosfomycin/pharmacology , Meropenem/pharmacology , Acinetobacter Infections/drug therapy , Anti-Bacterial Agents/administration & dosage , Carbapenems/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Fosfomycin/administration & dosage , Fosfomycin/pharmacokinetics , Meropenem/administration & dosage , Meropenem/pharmacokinetics , Microbial Sensitivity Tests , Monte Carlo Method
8.
J Chromatogr Sci ; 59(2): 165-174, 2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33302294

ABSTRACT

Fosfomycin is an antibiotic with a broad spectrum of activity against many multidrug-resistant bacterial strains. It is mainly excreted unchanged by the kidneys, and its half-life therefore depends on kidney function which varies considerably among individuals, and within individuals over time. Proper fosfomycin dosing thus depends on assaying blood concentration of the drug. We developed and validated a simple, sensitive and specific chromatography assay, which was coupled to electrospray ionization mass spectrometry for determination of fosfomycin. Separation of fosfomycin was based on the method of the hydrophilic interaction liquid chromatography; specifically, plasma and dialysate samples were acidified and the protein precipitated with acetonitrile. The calibration curves showed excellent coefficients of determination (R2 > 0.999) over the relevant concentration range of 25-700 µg/mL. Intraday precision was 1.1-1.2% and accuracy was -5.9% to 0.9% for quality control samples. Interday precision was 2.9-3.4% and accuracy was -3.7% to 5.5%. Extraction recovery was ≥87% and matrix effects ranged from 2.2% to 4.3%. After laboratory validation, the method was successfully applied to clinical samples.


Subject(s)
Chromatography, Liquid/methods , Fosfomycin/blood , Spectrometry, Mass, Electrospray Ionization/methods , Dialysis Solutions , Drug Stability , Fosfomycin/analysis , Fosfomycin/chemistry , Fosfomycin/pharmacokinetics , Humans , Hydrophobic and Hydrophilic Interactions , Limit of Detection , Linear Models , Reproducibility of Results , Sensitivity and Specificity
9.
J Chem Theory Comput ; 17(1): 549-559, 2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33378186

ABSTRACT

To reach their target site inside Gram-negative bacteria, almost all antibiotics need to cross the outer membrane. Computational modeling of such processes can be numerically demanding due to the size of the systems and especially due to the timescales involved. Recently, a hybrid Brownian and molecular dynamics approach, i.e., Brownian dynamics including explicit atoms (BRODEA), has been developed and evaluated for studying the transport of monoatomic ions through membrane channels. Later on, this numerically efficient scheme has been applied to determine the free energy surfaces of the ciprofloxacin and enrofloxacin translocation through the porin OmpC using temperature-accelerated simulations. To improve the usability and accuracy of the approach, schemes to approximate the position-dependent diffusion constant of the molecule while traversing the pore had to be established. To this end, we have studied the translocation of the charged phosphonic acid antibiotic fosfomycin through the porin OmpF from Escherichia coli devising and benchmarking several diffusion models. To test the efficiency and sensitivity of these models, the effect of OmpF mutations on the permeation of fosfomycin was analyzed. Permeation events have been recorded over millisecond-long biased and unbiased simulations, from which thermodynamics and kinetics quantities of the translocation processes were determined. As a result, the use of the BRODEA approach, together with the appropriate diffusion model, was seen to accurately reproduce the findings observed in electrophysiology experiments and all-atom molecular dynamics simulations. These results suggest that the BRODEA approach can become a valuable tool for screening numerous compounds to evaluate their outer membrane permeability, a property important in the development of new antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Escherichia coli/metabolism , Fosfomycin/pharmacokinetics , Porins/metabolism , Anti-Bacterial Agents/metabolism , Cell Membrane Permeability , Diffusion , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Infections/microbiology , Fosfomycin/metabolism , Humans , Kinetics , Models, Molecular , Mutation , Porins/chemistry , Porins/genetics , Protein Conformation , Thermodynamics
10.
Expert Rev Anti Infect Ther ; 18(7): 657-668, 2020 07.
Article in English | MEDLINE | ID: mdl-32397774

ABSTRACT

INTRODUCTION: Central nervous system (CNS) infections have considerable morbidity and mortality. Fosfomycin is a broad spectrum bactericidal antibiotic with favorable pharmacokinetic properties and low toxicity, satisfactory penetration in the cerebrospinal fluid and is authorized for the treatment of bacterial meningitis. AREAS COVERED: The objective of this analysis was to evaluate the available data regarding the effectiveness and safety of intravenous fosfomycin for the treatment of CNS infections. Thirty-two relevant publications were identified. Data from 224 patients who received intravenous fosfomycin as treatment for CNS infections were evaluated. Overall, 93.8% of patients were cured from the infection. Staphylococcus was the most frequent pathogen; Streptococcus pneumoniae, Neisseria meningitidis, and several other microbial agents, including multi-drug resistant and extensively drug-resistant bacteria, were also implicated. Fosfomycin was given as part of a combination treatment in the vast majority of the patients. The dosage of fosfomycin ranged between 4 g and 24 g per day; a regimen with 14-16 g per day was used in the majority of the cases. Fosfomycin was generally well tolerated. EXPERT OPINION: The evaluation of the published evidence suggests that fosfomycin may be beneficial in the treatment of patients with CNS infections.


Subject(s)
Bacterial Infections/drug therapy , Central Nervous System Infections/drug therapy , Fosfomycin/administration & dosage , Administration, Intravenous , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacokinetics , Bacterial Infections/microbiology , Central Nervous System Infections/microbiology , Dose-Response Relationship, Drug , Drug Resistance, Multiple, Bacterial , Fosfomycin/adverse effects , Fosfomycin/pharmacokinetics , Humans , Meningitis, Bacterial/drug therapy , Meningitis, Bacterial/microbiology , Treatment Outcome
11.
J Microbiol Methods ; 171: 105861, 2020 04.
Article in English | MEDLINE | ID: mdl-32035114

ABSTRACT

The impact of the bladder environment on fosfomycin activity and treatment response is uncertain. Standard laboratory media does not reflect the biomatrix of urine, where limited nutritional factors are important for growth and antimicrobial kill rates. We compared fosfomycin activity against Enterobacteriaceae in laboratory media, human urine and synthetic alternatives. Sixteen clinical isolates (8-Escherichia coli, 4-Enterobacter cloacae, 4-Klebsiella pneumoniae) were studied with broth microdilution (BMD) susceptibility, static time-kill assays and dynamic testing in a bladder infection model simulating a 3 g oral fosfomycin dose. Mueller-Hinton broth (MHB) with and without 25 mg/L glucose-6-phosphate (G6P), pooled midstream urine (MSU), pooled 24 h urine collection (24 U), artificial urine medium (AUM) and synthetic human urine (SHU) were compared. BMD susceptibility, bacterial growth and response to static fosfomycin concentrations in urine were best matched with SHU and were distinctly different when tested in MHB with G6P. Fosfomycin exposure in the bladder infection model was accurately reproduced (bias 4.7 ± 6.2%). Under all media conditions, 8 isolates (2-E. coli, 2-E. cloacae, 4-K. pneumoniae) re-grew and 4 isolates (4-E. coli) were killed. The remaining isolates (2-E. coli, 2-E. cloacae) re-grew variably in urine and synthetic media. Agar dilution MIC failed to predict re-growth, whereas BMD MIC in media without G6P performed better. Emergence of resistance was restricted in synthetic media. Overall, SHU provided the best substitute for urine for in vitro modelling of antimicrobial treatment of uropathogens, and these data have broader utility for improved preclinical testing of antimicrobials for urinary tract infections.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Enterobacter cloacae/drug effects , Escherichia coli/drug effects , Fosfomycin/pharmacokinetics , Klebsiella pneumoniae/drug effects , Urinary Tract Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/urine , Culture Media/chemistry , Drug Resistance, Bacterial/physiology , Enterobacter cloacae/isolation & purification , Escherichia coli/isolation & purification , Fosfomycin/pharmacology , Fosfomycin/urine , Humans , Klebsiella pneumoniae/isolation & purification , Microbial Sensitivity Tests , Urinary Bladder/microbiology , Urinary Bladder/pathology , Urinary Tract Infections/microbiology , Urine/microbiology
12.
Fundam Clin Pharmacol ; 34(4): 504-512, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31944378

ABSTRACT

We aimed to investigate the pharmacokinetics of fosfomycin and metronidazole after intraperitoneal administration of the combination of fosfomycin and metronidazole in patients undergoing laparoscopic appendectomy for uncomplicated appendicitis. We included eight otherwise healthy men undergoing laparoscopic appendectomy. The trial treatment was administered at the end of the surgical procedure and left in the abdominal cavity. Trial drugs consisted of 4 g fosfomycin and 1 g metronidazole in a total volume of 500.2 mL. Blood samples were collected prior to and ½, 1, 2, 4, 8, 12 and 24 h after administration. High-performance liquid chromatography-mass spectrometry was used for the measurement of plasma concentrations, and pharmacokinetic calculations were undertaken. Antimicrobial susceptibility testing was undertaken on isolates from intraoperatively collected specimens. The median maximal concentration for fosfomycin in plasma was 104.4 mg/L, median time point for the maximal concentration was 1.5 h, median half-life 3.0 h, and median area under the curve 608 mg*h/L. The median maximal concentration for metronidazole in plasma was 13.6 mg/L, median time point for the maximal concentration was 2.0 h, median half-life 7.3 h, and median area under the curve was 164 mg*h/L. All aerobic bacteria were susceptible to fosfomycin, and all anaerobes were susceptible to metronidazole. Plasma concentrations of fosfomycin and metronidazole were in line with concentrations reported from pharmacokinetic studies after intravenous administration and were within therapeutic ranges.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Appendectomy , Appendicitis/surgery , Fosfomycin/pharmacokinetics , Laparoscopy , Metronidazole/pharmacokinetics , Models, Biological , Postoperative Care , Surgical Wound Infection/prevention & control , Adolescent , Adult , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/blood , Appendectomy/adverse effects , Denmark , Drug Combinations , Drug Monitoring , Fosfomycin/administration & dosage , Fosfomycin/blood , Humans , Laparoscopy/adverse effects , Male , Metronidazole/administration & dosage , Metronidazole/blood , Middle Aged , Prospective Studies , Surgical Wound Infection/microbiology , Young Adult
13.
Int J Antimicrob Agents ; 55(4): 105861, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31838036

ABSTRACT

Antibiotic resistance is one of the major threats to public health today. To address this problem requires an urgent comprehensive approach. Strategic and multitargeted combination therapy has been increasingly used clinically to treat bacterial infections. The hollow-fibre infection model (HFIM) is a well-controlled in vitro bioreactor system that is increasingly being used in the assessment of resistance emergence with monotherapies and combination antibiotic therapies. In this study, the HFIM was evaluated as a reliable in vitro method to quantitatively and reproducibly analyse the emergence of antibiotic resistance using ampicillin, fosfomycin and ciprofloxacin and their simultaneous combinations against Escherichia coli CFT073, a clinical uropathogenic strain. Bacteria were exposed to clinically relevant pharmacokinetic (PK) concentrations of the drugs for 10 days. Drug and bacterial samples were collected at different time points for PK analysis and to enumerate total and resistant bacterial populations, respectively. The results demonstrated that double or triple combinations significantly delayed the emergence of resistant E. coli CFT073 subpopulations. These findings suggest that strategic combinations of antimicrobials may play a role in controlling the emergence of resistance during treatment. Further animal and human trials will be needed to confirm this and to ensure that there is no adverse impact on the host microbiome or unexpected toxicity. The HFIM system could potentially be used to identify clinically relevant combination dosing regimens for use in a clinical trial evaluating the appearance of resistance to antibacterial drugs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/pharmacokinetics , Bioreactors/microbiology , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/drug effects , Ampicillin/pharmacokinetics , Ampicillin/pharmacology , Ciprofloxacin/pharmacokinetics , Ciprofloxacin/pharmacology , Drug Combinations , Escherichia coli/genetics , Fosfomycin/pharmacokinetics , Fosfomycin/pharmacology , Humans , Microbial Sensitivity Tests
14.
Int J Antimicrob Agents ; 54(4): 435-441, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31382030

ABSTRACT

Given emerging uropathogen resistance to more recent antibiotics, old antibiotics used for uncomplicated urinary tract infection (UTI) warrant re-examination. In this study, the urinary antibacterial activities of fosfomycin and nitrofurantoin were investigated by determining the urinary inhibitory titre and urinary bactericidal titre against uropathogens in urine samples from female volunteers following administration of single-dose fosfomycin (3 g) or nitrofurantoin (50 mg q6h or 100 mg q8h). Urine samples were collected over 48 h (fosfomycin) or 6 or 8 h (nitrofurantoin), with drug levels quantified with every void. Fosfomycin concentrations ranged from <0.75 mg/L [lower limit of quantification (LLOQ)] to 5729.9 mg/L and nitrofurantoin concentrations ranged from <4 mg/L (LLOQ) to 176.3 mg/L (50 mg q6h) or 209.4 mg/L (100 mg q8h). There was discrepancy in the response to fosfomycin between Escherichia coli and Klebsiella pneumoniae, with fosfomycin displaying strong bactericidal activity for 48 h against E. coli but moderate bactericidal activity for 18 h against K. pneumoniae. This effect was not related to the strain's baseline minimum inhibitory concentration but rather to the presence of a resistant subpopulation. Maximum titres of nitrofurantoin were obtained during the first 2 h, but no antibacterial effect was found in most samples regardless of the dose. In the rare samples in which antibacterial activity was detectable, titres were comparable for both species tested. These findings confirm doubts regarding fosfomycin administration in UTIs caused by K. pneumoniae and reveal a discrepancy between nitrofurantoin's measurable ex vivo activity and its clinical effect over multiple dosing intervals.


Subject(s)
Anti-Infective Agents, Urinary/pharmacology , Anti-Infective Agents, Urinary/pharmacokinetics , Fosfomycin/pharmacology , Fosfomycin/pharmacokinetics , Nitrofurantoin/pharmacology , Nitrofurantoin/pharmacokinetics , Urine/chemistry , Adult , Anti-Infective Agents, Urinary/administration & dosage , Escherichia coli/drug effects , Female , Fosfomycin/administration & dosage , Healthy Volunteers , Humans , Klebsiella pneumoniae/drug effects , Microbial Sensitivity Tests , Microbial Viability/drug effects , Nitrofurantoin/administration & dosage , Urine/microbiology , Young Adult
15.
Indian J Med Microbiol ; 37(1): 95-98, 2019.
Article in English | MEDLINE | ID: mdl-31424016

ABSTRACT

There is a need of a relatively simple and inexpensive method for the determination of relative potency of various generic brands of antibiotics in comparison to original products. The current study describes an agar diffusion method which can be performed in any microbiology laboratory, is cheap (costs $2 per test) and its results can be available after overnight incubation. The results show that neither all generics are reliable nor are all generic antibiotics of poor quality.


Subject(s)
Anti-Bacterial Agents/pharmacology , Colistin/pharmacology , Drugs, Generic/pharmacology , Fosfomycin/pharmacology , Meropenem/pharmacology , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacokinetics , Bacteria/drug effects , Colistin/adverse effects , Colistin/pharmacokinetics , Drug Resistance, Multiple, Bacterial , Drugs, Generic/adverse effects , Drugs, Generic/pharmacokinetics , Fosfomycin/adverse effects , Fosfomycin/pharmacokinetics , Humans , Meropenem/adverse effects , Meropenem/pharmacokinetics , Microbial Sensitivity Tests , Therapeutic Equivalency
16.
J Antimicrob Chemother ; 74(8): 2335-2340, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31086958

ABSTRACT

OBJECTIVES: To assess the pharmacokinetics and tissue penetration of fosfomycin in obese and non-obese surgical patients. METHODS: Fifteen obese patients undergoing bariatric surgery and 15 non-obese patients undergoing major intra-abdominal surgery received an intravenous single short infusion of 8 g of fosfomycin. Fosfomycin concentrations were determined by LC-MS/MS in plasma and microdialysate from subcutaneous tissue up to 8 h after dosing. The pharmacokinetic analysis was performed in plasma and interstitial fluid (ISF) by non-compartmental methods. RESULTS: Thirteen obese patients (BMI 38-50 kg/m2) and 14 non-obese patients (BMI 0-29 kg/m2) were evaluable. The pharmacokinetics of fosfomycin in obese versus non-obese patients were characterized by lower peak plasma concentrations (468 ±âŸ139 versus 594 ±âŸ149 mg/L, P = 0.040) and higher V (24.4 ±âŸ6.4 versus 19.0 ±âŸ3.1 L, P = 0.010). The differences in AUC∞ were not significant (1275 ±âŸ477 versus 1515 ±âŸ352 mg·h/L, P = 0.16). The peak concentrations in subcutaneous tissue were reached rapidly and declined in parallel with the plasma concentrations. The drug exposure in tissue was nearly halved in obese compared with non-obese patients (AUC∞ 1052 ±âŸ394 versus 1929 ±âŸ725 mg·h/L, P = 0.0010). The tissue/plasma ratio (AUCISF/AUCplasma) was 0.86 ±âŸ0.32 versus 1.27 ±âŸ0.34 (P = 0.0047). CONCLUSIONS: Whereas the pharmacokinetics of fosfomycin in plasma of surgical patients were only marginally different between obese and non-obese patients, the drug exposure in subcutaneous tissue was significantly lower in the obese patients.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Fosfomycin/pharmacokinetics , Obesity , Plasma/chemistry , Subcutaneous Fat/chemistry , Adult , Aged , Anti-Bacterial Agents/administration & dosage , Chromatography, Liquid , Female , Fosfomycin/administration & dosage , Humans , Male , Middle Aged , Prospective Studies , Tandem Mass Spectrometry
17.
Rev Esp Quimioter ; 32 Suppl 1: 19-24, 2019 May.
Article in English | MEDLINE | ID: mdl-31131588

ABSTRACT

Fosfomycin, a low molecular weight and hydrophilic drug with negligible protein binding, is eliminated almost exclusively by glomerular filtration, whose clearance is subject to patient renal function. The volume of distribution approximates to the extracellular body water (about 0.3 L/Kg) in healthy volunteers, but it is increased in critically ill patients with bacterial infections. Fosfomycin presents a high ability to distribute into many tissues, including inflamed tissues and abscess fluids. Based on PK/PD analysis and Monte Carlo simulations, we have evaluated different fosfomycin dosing regimen to optimize the treatment of septic patients due to Enterobacterales and Pseudomonas aeruginosa. As PK/PD targets, we selected %T>MIC > 70% for all pathogens, and AUC24/MIC > 24 and AUC24/MIC > 15 for net stasis of Enterobacterales and P. aeruginosa, respectively. Pharmacokinetic parameters in critically ill patients were obtained from the literature. Several dosing regimens were studied in patients with normal renal function: fosfomycin 2-8 g given every 6-12 hours, infused over 30 minutes- 24 hours. At the susceptibility EUCAST breakpoint for Enterobacterales and Staphylococcus spp. (MIC ≤ 32 mg/L), fosfomycin 4 g/8h or higher infused over 30 minutes achieved a probability of target attainment (PTA) > 90%, based in both %T>MIC and AUC24/MIC. For MIC of 64 mg/L, fosfomycin 6 g/6h in 30-minute infusion and 8 g/ 8h in 30-minute and 6 hours infusions also achieved PTA values higher than 90%. No fosfomycin monotherapy regimen was able to achieve PK/PD targets related to antimicrobial efficacy for P. aeruginosa with MICs of 256-512 mg/L.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/pharmacokinetics , Fosfomycin/pharmacology , Fosfomycin/pharmacokinetics , Animals , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Fosfomycin/therapeutic use , Humans
18.
Rev Esp Quimioter ; 32 Suppl 1: 30-36, 2019 May.
Article in English | MEDLINE | ID: mdl-31131590

ABSTRACT

Osteoarticular infections include septic arthritis and osteomyelitis, with Gram-positive microorganisms isolated most frequently. In recent years, there has been an increase in the number of resistant strains in this type of infection, which complicates the treatment. Fosfomycin is active against a large percentage of Gram-positive and Gram-negative pathogens, including multidrug-resistant strains, and its properties include low protein binding, low molecular weight and good bone dissemination. In this article, we discuss fosfomycin's activity in vitro, its pharmacokinetic and pharmacodynamic parameters of interest in osteoarticular infections, the experimental models of osteomyelitis and foreign body infection and the clinical experience with these types of infections.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bone Diseases, Infectious/drug therapy , Cartilage Diseases/drug therapy , Fosfomycin/therapeutic use , Animals , Anti-Bacterial Agents/pharmacokinetics , Bone Diseases, Infectious/microbiology , Cartilage Diseases/microbiology , Fosfomycin/pharmacokinetics , Humans
19.
Rev Esp Quimioter ; 32 Suppl 1: 37-44, 2019 May.
Article in English | MEDLINE | ID: mdl-31131591

ABSTRACT

Urinary tract infections are one of the most common health problems and entail a high consumption of health system resources. Due to the increase in global antibiotic resistances in recent years, it is increasingly common to find uropathogens with multiple resistance mechanisms, including quinolone-resistant bacteria, broad-spectrum ß-lactamase producers and carbapenemase producers. In this scenario, the role of fosfomycin has gained considerable importance, given its spectrum of activity against multidrug resistant microorganisms (Gram-positive and Gram-negative), becoming an attractive alternative therapy. Regarding the use of fosfomycin in complicated urinary tract infections, there is increasing clinical experience with patients with infections caused by multidrug resistant bacteria, those with recurrent urinary tract infection and special populations such as those with kidney transplants. Randomized comparative studies and series are underway, which will provide greater evidence. Nevertheless, more studies are needed to confirm the enormous potential of fosfomycin in complicated urinary tract infection in the era of multiresistance.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Fosfomycin/therapeutic use , Urinary Tract Infections/drug therapy , Administration, Intravenous , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Drug Resistance, Multiple, Bacterial , Fosfomycin/administration & dosage , Fosfomycin/pharmacokinetics , Humans , Urinary Tract/metabolism , Urinary Tract Infections/complications , Urinary Tract Infections/microbiology
20.
Rev. esp. quimioter ; 32(supl.1): 19-24, mayo 2019. tab
Article in English | IBECS | ID: ibc-188722

ABSTRACT

Fosfomycin, a low molecular weight and hydrophilic drug with negligible protein binding, is eliminated almost exclusively by glomerular filtration, whose clearance is subject to patient renal function. The volume of distribution approximates to the extracellular body water (about 0.3 L/Kg) in healthy volunteers, but it is increased in critically ill patients with bacterial infections. Fosfomycin presents a high ability to distribute into many tissues, including inflamed tissues and abscess fluids. Based on PK/PD analysis and Monte Carlo simulations, we have evaluated different fosfomycin dosing regimen to optimize the treatment of septic patients due to Enterobacterales and Pseudomonas aeruginosa. As PK/PD targets, we selected %T>MIC > 70% for all pathogens, and AUC24/MIC > 24 and AUC24/MIC > 15 for net stasis of Enterobacterales and P. aeruginosa, respectively. Pharmacokinetic parameters in critically ill patients were obtained from the literature. Several dosing regimens were studied in patients with normal renal function: fosfomycin 2-8 g given every 6-12 hours, infused over 30 minutes- 24 hours. At the susceptibility EUCAST breakpoint for Enterobacterales and Staphylococcus spp. (MIC ≤ 32 mg/L), fosfomycin 4 g/8h or higher infused over 30 minutes achieved a probability of target attainment (PTA) > 90%, based in both %T>MIC and AUC24/MIC. For MIC of 64 mg/L, fosfomycin 6 g/6h in 30-minute infusion and 8 g/ 8h in 30-minute and 6 hours infusions also achieved PTA values higher than 90%. No fosfomycin monotherapy regimen was able to achieve PK/PD targets related to antimicrobial efficacy for P. aeruginosa with MICs of 256-512 mg/L


Fosfomicina, fármaco de bajo peso molecular e hidrófilo con insignificante unión a proteínas, se elimina casi exclusivamente por filtración glomerular, y su aclaramiento está condicionado por la función renal del paciente. El volumen de distribución se asemeja al agua corporal extracelular (aproximadamente 0,3 L/Kg) en voluntarios sanos, pero es mayor en pacientes con infecciones bacterianas graves. Fosfomicina presenta una alta capacidad de distribución a una gran variedad de tejidos, incluidos tejidos inflamados y abscesos. Mediante análisis PK/PD y simulación de Montecarlo, hemos evaluado distintas pautas posológicas de fosfomicina para optimizar el tratamiento de pacientes sépticos debido a Enterobacterales y Pseudomonas aeruginosa.como objetivos PK/PD, seleccionamos %T>MIC >70% para todos los patógenos, y AUC24/CMI >24 y AUC24/CMI >15 para estasis neta de Enterobacterales y P. aeruginosa, respectivamente. Los parámetros farmacocinéticos en pacientes críticos se obtuvieron de la literatura. Se estudiaron diversas pautas posológicas en pacientes con función renal normal: de 2 a 8 g de fosfomicina cada 6 a 12 h, en perfusión de entre 30 minutos y 24 horas. En el punto de corte de sensibilidad de EUCAST para Enterobacterales y Staphylococcus spp. (CMI ≤32 mg/L), con 4 g/8 h o más de fosfomicina en perfusión de 30 minutos se alcanzó una probabilidad de alcanzar el objetivo terapéutico (PTA) del 90%, considerando tanto %T>CMI como AUC24/CMI. Para la CMI de 64 mg/L, 6 g/6 h de fosfomicina en infusión de 30 minutos y 8 g/8 h en infusión de entre 30 minutos y 6 horas también proporcionaron valores de PTA superiores al 90%. Ninguna pauta con monoterapia de fosfomicina logró alcanzar los objetivos PK/PD en relación con la eficacia antimicrobiana para P. aeruginosa con CMI de 256 y 512 mg/L


Subject(s)
Humans , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/pharmacokinetics , Fosfomycin/pharmacology , Fosfomycin/pharmacokinetics , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Fosfomycin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL