Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Diagn Microbiol Infect Dis ; 109(3): 116344, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38735147

RESUMEN

Combinations of the ß-lactam/ß-lactamase inhibitor sulbactam-durlobactam and seventeen antimicrobial agents were tested against strains of Acinetobacter baumannii in checkerboard assays. Most combinations resulted in indifference with no instances of antagonism. These results suggest sulbactam-durlobactam antibacterial activity against A. baumannii is unlikely to be affected if co-dosed with other antimicrobial agents.


Asunto(s)
Acinetobacter baumannii , Antibacterianos , Compuestos de Azabiciclo , Pruebas de Sensibilidad Microbiana , Sulbactam , Sulbactam/farmacología , Acinetobacter baumannii/efectos de los fármacos , Compuestos de Azabiciclo/farmacología , Antibacterianos/farmacología , Humanos , Acinetobacter calcoaceticus/efectos de los fármacos , Inhibidores de beta-Lactamasas/farmacología , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/tratamiento farmacológico , Combinación de Medicamentos
2.
J Clin Microbiol ; 62(1): e0122823, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-38095417

RESUMEN

Sulbactam-durlobactam is a ß-lactam/ß-lactamase inhibitor combination developed to treat hospital-acquired and ventilator-associated bacterial pneumonia caused by Acinetobacter baumannii-calcoaceticus complex (ABC). Durlobactam is a diazabicyclooctane ß-lactamase inhibitor with potent activity against Ambler classes A, C, and D serine ß-lactamases and restores sulbactam activity against multidrug-resistant ABC. Studies were conducted to establish sulbactam-durlobactam antimicrobial susceptibility testing methods for both broth microdilution minimal inhibitory concentration (MIC) and disk diffusion tests as well as quality control (QC) ranges. To establish the MIC test method, combinations of sulbactam and durlobactam were evaluated using a panel of genetically characterized A. baumannii isolates which were categorized as predicted to be susceptible or resistant based on the spectrum of ß-lactamase inhibition by durlobactam. MIC testing with doubling dilutions of sulbactam with a fixed concentration of 4 µg/mL of durlobactam resulted in the greatest discrimination of the pre-defined susceptible and resistant strains. Similarly, the sulbactam/durlobactam 10/10 µg disk concentration showed the best discrimination as well as correlation with the MIC test. A. baumannii NCTC 13304 was selected for QC purposes because it assesses the activity of both sulbactam and durlobactam with clear endpoints. Multi-laboratory QC studies were conducted according to CLSI M23 Tier 2 criteria. A sulbactam-durlobactam broth MIC QC range of 0.5/4-2/4 µg/mL and a zone diameter QC range of 24-30 mm were determined for A. baumannii NCTC 13304 and have been approved by CLSI. These studies will enable clinical laboratories to perform susceptibility tests with accurate and reproducible methods.


Asunto(s)
Acinetobacter baumannii , Compuestos de Azabiciclo , Sulbactam , Humanos , Sulbactam/farmacología , Sulbactam/uso terapéutico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Inhibidores de beta-Lactamasas/farmacología , Inhibidores de beta-Lactamasas/uso terapéutico , Pruebas de Sensibilidad Microbiana , Control de Calidad , Combinación de Medicamentos
3.
J Antimicrob Chemother ; 78(11): 2672-2682, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37700689

RESUMEN

OBJECTIVES: This post hoc pooled analysis evaluated clinical and microbiological outcomes and safety in patients with infections caused by ß-lactamase-producing Gram-negative pathogens across five Phase 3, randomized, controlled, multicentre trials of ceftazidime/avibactam in adults with complicated intra-abdominal infection (cIAI), complicated urinary tract infection (cUTI)/pyelonephritis and nosocomial pneumonia (NP), including ventilator-associated pneumonia (VAP). METHODS: In each trial, RECLAIM/RECLAIM 3 (cIAI), REPRISE (cIAI/cUTI), RECAPTURE (cUTI) and REPROVE (NP, including VAP) patients were randomized 1:1 to IV ceftazidime/avibactam (plus metronidazole for patients with cIAI) or comparators (carbapenems in >97% patients) for 5-21 days. Clinical and microbiological responses at the test-of-cure visit were assessed for patients with ESBLs, and/or plasmidic and/or overexpression of chromosomal AmpC, and/or serine carbapenemases without MBLs identified in baseline Gram-negative isolates by phenotypic screening and molecular characterization in the pooled microbiological modified ITT (mMITT) population. RESULTS: In total, 813 patients (ceftazidime/avibactam, n = 389; comparator, n = 424) had ≥1 ß-lactamase-producing baseline pathogen identified, amongst whom 792 patients (ceftazidime/avibactam, n = 379; comparator, n = 413) had no MBLs. The most frequent ß-lactamase-producing pathogens across treatment groups were Escherichia coli (n = 381), Klebsiella pneumoniae (n = 261) and Pseudomonas aeruginosa (n = 53). Clinical cure rates in the pooled non-MBL ß-lactamase-producing mMITT population were 88.1% (334/379) for ceftazidime/avibactam and 88.1% (364/413) for comparators; favourable microbiological response rates were 76.5% (290/379) and 68.8% (284/413), respectively. The safety profile of ceftazidime/avibactam was consistent with previous observations. CONCLUSIONS: This analysis provides supportive evidence of the efficacy and safety of ceftazidime/avibactam in patients with infections caused by ESBLs, AmpC and serine carbapenemase-producing Gram-negative pathogens. TRIAL REGISTRATION: NCT01499290; NCT01726023; NCT01644643; NCT01595438/NCT01599806; NCT01808092.


Asunto(s)
Infecciones Intraabdominales , Infecciones Urinarias , Adulto , Humanos , Antibacterianos/efectos adversos , beta-Lactamasas , Ceftazidima/efectos adversos , Escherichia coli , Infecciones Intraabdominales/tratamiento farmacológico , Infecciones Intraabdominales/microbiología , Ensayos Clínicos Controlados Aleatorios como Asunto , Serina/uso terapéutico , Infecciones Urinarias/microbiología
4.
JAC Antimicrob Resist ; 5(4): dlad097, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37583473

RESUMEN

Antimicrobial resistance (AMR) continues to serve as a major global health crisis. Clinicians practising in this modern era are faced with ongoing challenges in the therapeutic management of patients suffering from antimicrobial-resistant infections. A strong educational understanding and synergistic application of clinical microbiology, infectious disease and pharmacological concepts can assist the adventuring clinician in the navigation of such cases. Important items include mobilizing laboratory testing for pathogen identification and susceptibility data, harnessing an understanding of intrinsic pathogen resistance, acknowledging epidemiological resistance trends, recognizing acquired AMR mechanisms, and consolidating these considerations when constructing an ideal pharmacological plan. In this article, we outline a novel framework by which to systematically approach clinical AMR, encourage AMR-related education and optimize therapeutic decision-making in AMR-related illnesses.

5.
J Antimicrob Chemother ; 78(4): 871-892, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36883356

RESUMEN

As one of a series of thematically linked reviews of the primary pharmacology of the ß-lactam/ß-lactamase inhibitor combination, ceftazidime/avibactam, this article reviews the microbiological findings in drug-exposed patients. Earlier articles in the series focused on basic in vitro and in vivo translational biology (J Antimicrob Chemother 2022; 77: 2321-40 and 2341-52) and the development and mechanisms of resistance in vitro (J Antimicrob Chemother 2023: Epub ahead of print. doi: 10.1093/jac/dkac449). In clinical trials of ceftazidime/avibactam, combined favourable microbiological responses for evaluable patients infected at baseline by susceptible Enterobacterales or Pseudomonas aeruginosa were 86.1% (851/988). The corresponding percent favourable among patients infected by ceftazidime/avibactam-resistant pathogens was 58.8% (10/17), noting that the majority (15/17) of the resistant examples were P. aeruginosa. Microbiological response rates to comparator treatments in the same clinical trials ranged between 64% and 95%, depending on the type of infection and the analysis population. Uncontrolled case studies over a wide range of patients infected by antibiotic multiresistant Gram-negative bacteria have demonstrated that ceftazidime/avibactam can elicit microbiological clearance of ceftazidime/avibactam-susceptible strains. In case studies where a matched cohort of patients had been treated with antibacterial agents other than ceftazidime/avibactam, microbiological outcomes were comparable between treatments, mostly being observationally more favourable for ceftazidime/avibactam (recognizing that numbers were too small for definitive superiority assessments). Development of resistance to ceftazidime/avibactam during therapy is reviewed. The phenomenon has been reported multiple times, mostly in difficult-to-treat patients infected by KPC-producing Enterobacterales. Molecular mechanisms, when determined, have frequently been observed previously in vitro, such as the 'Ω-loop' D179Y (Asp179Tyr) substitution found in KPC variant enzymes. In human volunteers exposed to therapeutic levels of ceftazidime/avibactam, faecal numbers of Escherichia coli, other enterobacteria, lactobacilli, bifidobacteria, clostridia and Bacteroides spp. decreased. Clostridioides difficile was detected in the faeces, but this was of uncertain significance, because no unexposed controls were studied.


Asunto(s)
Antibacterianos , Ceftazidima , Humanos , Ceftazidima/farmacología , Ceftazidima/uso terapéutico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Compuestos de Azabiciclo/farmacología , Compuestos de Azabiciclo/uso terapéutico , Inhibidores de beta-Lactamasas/farmacología , Inhibidores de beta-Lactamasas/uso terapéutico , Combinación de Medicamentos , Bacterias Gramnegativas , Escherichia coli , Pruebas de Sensibilidad Microbiana , beta-Lactamasas , Pseudomonas aeruginosa
6.
J Antimicrob Chemother ; 78(3): 569-585, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36702744

RESUMEN

This article reviews resistance to ceftazidime/avibactam as an aspect of its primary pharmacology, linked thematically with recent reviews of the basic in vitro and in vivo translational biology of the combination (J Antimicrob Chemother 2022; 77: 2321-40 and 2341-52). In Enterobacterales or Pseudomonas aeruginosa, single-step exposures to 8×  MIC of ceftazidime/avibactam yielded frequencies of resistance from <∼0.5 × 10-9 to 2-8 × 10-9, depending on the host strain and the ß-lactamase harboured. ß-Lactamase structural gene mutations mostly affected the avibactam binding site through changes in the Ω-loop: e.g. Asp179Tyr (D179Y) in KPC-2. Other mutations included ones proposed to reduce the permeability to ceftazidime and/or avibactam through changes in outer membrane structure, up-regulated efflux, or both. The existence, or otherwise, of cross-resistance between ceftazidime/avibactam and other antibacterial agents was also reviewed as a key element of the preclinical primary pharmacology of the new agent. Cross-resistance between ceftazidime/avibactam and other ß-lactam-based antibacterial agents was caused by MBLs. Mechanism-based cross-resistance was not observed between ceftazidime/avibactam and fluoroquinolones, aminoglycosides or colistin. A low level of general co-resistance to ceftazidime/avibactam was observed in MDR Enterobacterales and P. aeruginosa. For example, among 2821 MDR Klebsiella spp., 3.4% were resistant to ceftazidime/avibactam, in contrast to 0.07% of 8177 non-MDR isolates. Much of this was caused by possession of MBLs. Among 1151 MDR, XDR and pandrug-resistant isolates of P. aeruginosa from the USA, 11.1% were resistant to ceftazidime/avibactam, in contrast to 3.0% of 7452 unselected isolates. In this case, the decreased proportion susceptible was not due to MBLs.


Asunto(s)
Antibacterianos , Ceftazidima , Antibacterianos/farmacología , Compuestos de Azabiciclo/farmacología , beta-Lactamasas/genética , Ceftazidima/farmacología , Combinación de Medicamentos , Pruebas de Sensibilidad Microbiana , Pseudomonas aeruginosa/genética , Resistencia a Medicamentos
7.
J Antimicrob Chemother ; 77(9): 2321-2340, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-35665807

RESUMEN

Previous reviews of ceftazidime/avibactam have focused on in vitro molecular enzymology and microbiology or the clinically associated properties of the combination. Here we take a different approach. We initiate a series of linked reviews that analyse research on the combination that built the primary pharmacology data required to support the clinical and business risk decisions to perform randomized controlled Phase 3 clinical trials, and the additional microbiological research that was added to the above, and the safety and chemical manufacturing and controls data, that constituted successful regulatory licensing applications for ceftazidime/avibactam in multiple countries, including the USA and the EU. The aim of the series is to provide both a source of reference for clinicians and microbiologists to be able to use ceftazidime/avibactam to its best advantage for patients, but also a case study of bringing a novel ß-lactamase inhibitor (in combination with an established ß-lactam) through the microbiological aspects of clinical development and regulatory applications, updated finally with a review of resistance occurring in patients under treatment. This first article reviews the biochemistry, structural biology and basic microbiology of the combination, showing that avibactam inhibits the great majority of serine-dependent ß-lactamases in Enterobacterales and Pseudomonas aeruginosa to restore the in vitro antibacterial activity of ceftazidime. Translation to efficacy against infections in vivo is reviewed in the second co-published article, Nichols et al. (J Antimicrob Chemother 2022; 77: 2341-52).


Asunto(s)
Compuestos de Azabiciclo , Ceftazidima , Antibacterianos/uso terapéutico , Compuestos de Azabiciclo/uso terapéutico , Biología , Ceftazidima/uso terapéutico , Ensayos Clínicos Fase III como Asunto , Combinación de Medicamentos , Humanos , Pruebas de Sensibilidad Microbiana , Pseudomonas aeruginosa , Ensayos Clínicos Controlados Aleatorios como Asunto , Inhibidores de beta-Lactamasas/farmacología , Inhibidores de beta-Lactamasas/uso terapéutico , beta-Lactamasas
8.
J Antimicrob Chemother ; 77(9): 2341-2352, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-35660869

RESUMEN

This review describes the translational in vivo and non-clinical pharmacokinetics/pharmacodynamics (PK/PD) research that supported clinical trialling and subsequently licensing approval of ceftazidime/avibactam, a new ß-lactam/ß-lactamase inhibitor combination aimed at the treatment of infections by Enterobacterales and Pseudomonas aeruginosa. The review thematically follows on from the co-published article, Nichols et al. (J Antimicrob Chemother 2022; 77: 2321-40). Avibactam protected ceftazidime in animal models of infection with ceftazidime-resistant, ß-lactamase-producing bacteria. For example, a single subcutaneous dose of ceftazidime at 1024 mg/kg yielded little effect on the growth of ceftazidime-resistant, blaKPC-2-carrying Klebsiella pneumoniae in the thighs of neutropenic mice (final counts of 4 × 108 to 8 × 108 cfu/thigh). In contrast, co-administration of avibactam in a 4:1 ratio (ceftazidime:avibactam) was bactericidal in the same model (final counts of 2 × 104 to 3 × 104 cfu/thigh). In a rat abdominal abscess model, therapy with ceftazidime or ceftazidime/avibactam (4:1 w/w) against blaKPC-2-positive K. pneumoniae resulted in 9.3 versus 3.3 log cfu/abscess, respectively, after 52 h. With respect to PK/PD, in Monte Carlo simulations, attainment of unbound drug exposure targets (ceftazidime fT>8 mg/L and avibactam fT>1 mg/L, each for 50% of the dosing interval) for the labelled dose of ceftazidime/avibactam (2 and 0.5 g, respectively, q8h by 2 h IV infusion), including dose adjustments for patients with impaired renal function, ranged between 94.8% and 99.6% of patients, depending on the infection modelled.


Asunto(s)
Compuestos de Azabiciclo , Ceftazidima , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Compuestos de Azabiciclo/farmacología , Compuestos de Azabiciclo/uso terapéutico , Biología , Ceftazidima/farmacología , Ceftazidima/uso terapéutico , Combinación de Medicamentos , Klebsiella pneumoniae , Ratones , Pruebas de Sensibilidad Microbiana , Ratas , Inhibidores de beta-Lactamasas/farmacología , Inhibidores de beta-Lactamasas/uso terapéutico
10.
Antimicrob Agents Chemother ; 66(4): e0033322, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35380458

RESUMEN

Assigning names to ß-lactamase variants has been inconsistent and has led to confusion in the published literature. The common availability of whole genome sequencing has resulted in an exponential growth in the number of new ß-lactamase genes. In November 2021 an international group of ß-lactamase experts met virtually to develop a consensus for the way naturally-occurring ß-lactamase genes should be named. This document formalizes the process for naming novel ß-lactamases, followed by their subsequent publication.


Asunto(s)
Inhibidores de beta-Lactamasas , beta-Lactamasas , Consenso , beta-Lactamasas/genética
11.
JAC Antimicrob Resist ; 3(3): dlab092, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34286272

RESUMEN

Extended-spectrum ß-lactamase (ESBL)-producing Gram-negative pathogens are a major cause of resistance to expanded-spectrum ß-lactam antibiotics. Since their discovery in the early 1980s, they have spread worldwide and an are now endemic in Enterobacterales isolated from both hospital-associated and community-acquired infections. As a result, they are a global public health concern. In the past, TEM- and SHV-type ESBLs were the predominant families of ESBLs. Today CTX-M-type enzymes are the most commonly found ESBL type with the CTX-M-15 variant dominating worldwide, followed in prevalence by CTX-M-14, and CTX-M-27 is emerging in certain parts of the world. The genes encoding ESBLs are often found on plasmids and harboured within transposons or insertion sequences, which has enabled their spread. In addition, the population of ESBL-producing Escherichia coli is dominated globally by a highly virulent and successful clone belonging to ST131. Today, there are many diagnostic tools available to the clinical microbiology laboratory and include both phenotypic and genotypic tests to detect ß-lactamases. Unfortunately, when ESBLs are not identified in a timely manner, appropriate antimicrobial therapy is frequently delayed, resulting in poor clinical outcomes. Several analyses of clinical trials have shown mixed results with regards to whether a carbapenem must be used to treat serious infections caused by ESBLs or whether some of the older ß-lactam-ß-lactamase combinations such as piperacillin/tazobactam are appropriate. Some of the newer combinations such as ceftazidime/avibactam have demonstrated efficacy in patients. ESBL-producing Gram-negative pathogens will continue to be major contributor to antimicrobial resistance worldwide. It is essential that we remain vigilant about identifying them both in patient isolates and through surveillance studies.

12.
Eur J Clin Microbiol Infect Dis ; 39(8): 1559-1572, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32356026

RESUMEN

Antibiotic resistance of bacterial pathogens isolated in China is a major concern. Omadacycline is a novel tetracycline derivative that has been approved for use in skin infections and community-acquired pneumonia. This study was conducted to determine the in vitro activity of omadacycline against a large collection of patient isolate medical centers across Mainland China. A total of 1041 recent clinical isolates are obtained from patients hospitalized in 29 provinces and municipalities across China. The in vitro activity of omadacycline and comparator agents was assessed using the microbroth dilution methodology. Omadacycline was active against methicillin-susceptible and -resistant Staphylococcus aureus with MIC90 values of 0.25 and 1 mg/L, respectively. All isolates of Enterococcus faecalis and Enterococcus faecium, including vancomycin-resistant isolates, were inhibited by ≤ 0.25 mg/L of omadacycline. It was active against Streptococcus pneumoniae irrespective of susceptibility to penicillin or macrolides (MIC90 =0.12 mg/L). The minimum inhibitory concentration (MIC) distribution of omadacycline was nearly identical against (extended-spectrum beta-lactamases) ESBL-positive, ESBL-negative, and carbapenemase-producing Escherichia coli (MIC90 = 4 mg/L). Omadacycline also showed good activity against Acinetobacter baumannii, inhibiting all isolates at ≤ 8 mg/L. Against Hemophilus influenzae and Moraxella catarrhalis, the MICs of omadacycline were low and not influenced by the presence of ß-lactamase. Overall, the activity of omadacycline was very good against isolates commonly associated with skin infections and pneumonia, and the susceptibility of Chinese isolates was similar to that reported for these pathogens from large surveillance studies outside China. This suggests that omadacycline could be an option for treatment of these infections in Chinese patients.


Asunto(s)
Antibacterianos/farmacología , Infecciones Comunitarias Adquiridas/epidemiología , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Tetraciclinas/farmacología , Antibacterianos/uso terapéutico , China/epidemiología , Infecciones Comunitarias Adquiridas/tratamiento farmacológico , Infecciones Comunitarias Adquiridas/microbiología , Demografía , Farmacorresistencia Bacteriana Múltiple , Humanos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Tetraciclinas/uso terapéutico
13.
ACS Infect Dis ; 6(6): 1332-1345, 2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32329999

RESUMEN

The Centers for Disease Control and the World Health Organization have issued a list of priority pathogens for which there are dwindling therapeutic options, including antibiotic-resistant Neisseria gonorrheae, for which novel oral agents are urgently needed. Zoliflodacin, the first in a new class of antibacterial agents called the spiropyrimidinetriones, is being developed for the treatment of gonorrhea. It has a unique mode of inhibition against bacterial type II topoisomerases with binding sites in bacterial gyrase that are distinct from those of the fluoroquinolones. Zoliflodacin is bactericidal, with a low frequency of resistance and potent antibacterial activity against N. gonorrheae, including multi-drug-resistant strains (MICs ranging from ≤0.002 to 0.25 µg/mL). Although being developed for the treatment of gonorrhea, zoliflodacin also has activity against Gram-positive, fastidious Gram-negative, and atypical pathogens. A hollow-fiber infection model using S. aureus showed that that pharmacokinetic/pharmacodynamic index of fAUC/MIC best correlated with efficacy in in vivo neutropenic thigh models in mice. This data and unbound exposure magnitudes derived from the thigh models were subsequently utilized in a surrogate pathogen approach to establish dose ranges for clinical development with N. gonorrheae. In preclinical studies, a wide safety margin supported progression to phase 1 studies in healthy volunteers, which showed linear pharmacokinetics, good oral bioavailability, and no significant safety findings. In a phase 2 study, zoliflodacin was effective in treating gonococcal urogenital and rectal infections. In partnership with the Global Antibiotic Research Development Program (GARDP), zoliflodacin is currently being studied in a global phase 3 clinical trial. Zoliflodacin represents a promising new oral therapy for drug-resistant infections caused by N. gonorrheae.


Asunto(s)
Antibacterianos , Preparaciones Farmacéuticas , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Barbitúricos , Isoxazoles , Ratones , Morfolinas , Neisseria gonorrhoeae , Oxazolidinonas , Compuestos de Espiro , Staphylococcus aureus
14.
J Antimicrob Chemother ; 75(7): 1833-1839, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32306049

RESUMEN

BACKGROUND: Durlobactam is a broad-spectrum inhibitor of class A, C and D ß-lactamases. Sulbactam is a generic ß-lactam most commonly used as a ß-lactamase inhibitor in combination with ampicillin; however, it has a unique property in that it has selective intrinsic activity against Acinetobacter baumannii. Currently, there is widespread resistance caused by multiple ß-lactamases including class A carbapenemases and class C and class D enzymes. The addition of durlobactam to sulbactam restores in vitro activity against MDR A. baumannii that possess multiple ß-lactamases. OBJECTIVES: Previously, susceptibility data for sulbactam/durlobactam were limited to isolates from patients in Western countries. This study was undertaken to determine the activity of sulbactam/durlobactam against A. baumannii isolated from patients in mainland China. METHODS: Nine hundred and eighty-two recent A. baumannii clinical isolates were collected from 22 sites across mainland China during 2016-18. The isolates were collected from lower respiratory tract, intra-abdominal, urinary tract and skin and skin structure infections. The in vitro activities of sulbactam/durlobactam and comparators were determined by broth microdilution. RESULTS: The addition of durlobactam restored the activity of sulbactam against the majority of the strains tested. The MIC90 of sulbactam/durlobactam was 2 mg/L for all A. baumannii, compared with 64 mg/L for sulbactam alone. The MIC90 of sulbactam/durlobactam of 2 mg/L remained unchanged for 831 carbapenem-resistant isolates. Colistin was the only comparator with comparable activity (MIC90 = 1 mg/L). CONCLUSIONS: This study demonstrated the potential utility of sulbactam/durlobactam for the treatment of infections caused by A. baumannii in China.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Antibacterianos/farmacología , China , Farmacorresistencia Bacteriana Múltiple , Humanos , Pruebas de Sensibilidad Microbiana , Sulbactam/farmacología
15.
Artículo en Inglés | MEDLINE | ID: mdl-32071051

RESUMEN

Nosocomial pneumonia (NP), including ventilator-associated pneumonia (VAP), is increasingly associated with multidrug-resistant Gram-negative pathogens. This study describes the in vitro activity of ceftazidime-avibactam, ceftazidime, and relevant comparator agents against bacterial pathogens isolated from patients with NP, including VAP, enrolled in a ceftazidime-avibactam phase 3 trial. Gram-positive pathogens were included if coisolated with a Gram-negative pathogen. In vitro susceptibility was determined at a central laboratory using Clinical and Laboratory Standards Institute broth microdilution methods. Of 817 randomized patients, 457 (55.9%) had ≥1 Gram-negative bacterial pathogen(s) isolated at baseline, and 149 (18.2%) had ≥1 Gram-positive pathogen(s) coisolated. The most common isolated pathogens were Klebsiella pneumoniae (18.8%), Pseudomonas aeruginosa (15.8%), and Staphylococcus aureus (11.5%). Ceftazidime-avibactam was highly active in vitro against 370 isolates of Enterobacteriaceae, with 98.6% susceptible (MIC90, 0.5 µg/ml) compared with 73.2% susceptible for ceftazidime (MIC90, >64 µg/ml). The percent susceptibility values for ceftazidime-avibactam and ceftazidime against 129 P. aeruginosa isolates were 88.4% and 72.9% (MIC90 values of 16 µg/ml and 64 µg/ml), respectively. Among ceftazidime-nonsusceptible Gram-negative isolates, ceftazidime-avibactam percent susceptibility values were 94.9% for 99 Enterobacteriaceae and 60.0% for 35 P. aeruginosa MIC90 values for linezolid and vancomycin (permitted per protocol for Gram-positive coverage) were within their respective MIC susceptibility breakpoints against the Gram-positive pathogens isolated. This analysis demonstrates that ceftazidime-avibactam was active in vitro against the majority of Enterobacteriaceae and P. aeruginosa isolates from patients with NP, including VAP, in a phase 3 trial. (This study has been registered at ClinicalTrials.gov under identifier NCT01808092.).


Asunto(s)
Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Compuestos de Azabiciclo/farmacología , Compuestos de Azabiciclo/uso terapéutico , Ceftazidima/farmacología , Ceftazidima/uso terapéutico , Neumonía Asociada a la Atención Médica/tratamiento farmacológico , Neumonía Asociada a la Atención Médica/microbiología , Neumonía Asociada al Ventilador/tratamiento farmacológico , Neumonía Asociada al Ventilador/microbiología , Inhibidores de beta-Lactamasas/farmacología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Método Doble Ciego , Combinación de Medicamentos , Femenino , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Adulto Joven
16.
Clin Microbiol Rev ; 33(2)2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-32102899

RESUMEN

ß-Lactam antibiotics have been widely used as therapeutic agents for the past 70 years, resulting in emergence of an abundance of ß-lactam-inactivating ß-lactamases. Although penicillinases in Staphylococcus aureus challenged the initial uses of penicillin, ß-lactamases are most important in Gram-negative bacteria, particularly in enteric and nonfermentative pathogens, where collectively they confer resistance to all ß-lactam-containing antibiotics. Critical ß-lactamases are those enzymes whose genes are encoded on mobile elements that are transferable among species. Major ß-lactamase families include plasmid-mediated extended-spectrum ß-lactamases (ESBLs), AmpC cephalosporinases, and carbapenemases now appearing globally, with geographic preferences for specific variants. CTX-M enzymes include the most common ESBLs that are prevalent in all areas of the world. In contrast, KPC serine carbapenemases are present more frequently in the Americas, the Mediterranean countries, and China, whereas NDM metallo-ß-lactamases are more prevalent in the Indian subcontinent and Eastern Europe. As selective pressure from ß-lactam use continues, multiple ß-lactamases per organism are increasingly common, including pathogens carrying three different carbapenemase genes. These organisms may be spread throughout health care facilities as well as in the community, warranting close attention to increased infection control measures and stewardship of the ß-lactam-containing drugs in an effort to control selection of even more deleterious pathogens.


Asunto(s)
Bacterias/efectos de los fármacos , Infecciones Bacterianas/epidemiología , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana Múltiple , beta-Lactamasas/genética , beta-Lactamas/farmacología , Animales , Antibacterianos/farmacología , Bacterias/enzimología , Bacterias/genética , Infecciones Bacterianas/terapia , Técnicas de Laboratorio Clínico , Humanos , Plásmidos
17.
Artículo en Inglés | MEDLINE | ID: mdl-31712217

RESUMEN

Unlike for classes A and B, a standardized amino acid numbering scheme has not been proposed for the class C (AmpC) ß-lactamases, which complicates communication in the field. Here, we propose a scheme developed through a collaborative approach that considers both sequence and structure, preserves traditional numbering of catalytically important residues (Ser64, Lys67, Tyr150, and Lys315), is adaptable to new variants or enzymes yet to be discovered and includes a variation for genetic and epidemiological applications.


Asunto(s)
Proteínas Bacterianas/clasificación , Bacterias Gramnegativas/genética , Bacterias Grampositivas/genética , Mutación , Terminología como Asunto , Resistencia betalactámica/genética , beta-Lactamasas/clasificación , Secuencia de Aminoácidos , Antibacterianos/química , Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Expresión Génica , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/enzimología , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/enzimología , Cooperación Internacional , Estructura Secundaria de Proteína , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Inhibidores de beta-Lactamasas/química , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , beta-Lactamas/química , beta-Lactamas/farmacología
18.
J Clin Microbiol ; 57(9)2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31315953

RESUMEN

This report describes the results of two different, multilaboratory quality control (QC) studies that were used to establish QC ranges for the novel gyrase inhibitor zoliflodacin against the ATCC strains recommended by the Clinical and Laboratory Standards Institute (CLSI). Following the completion of an eight-laboratory, CLSI document M23-defined tier 2 study, the agar dilution MIC QC range for zoliflodacin against the Neisseria gonorrhoeae QC strain ATCC 49226 was defined as 0.06 to 0.5 µg/ml and was approved by the CLSI Subcommittee on Antimicrobial Susceptibility Testing. This QC range will be used for in vitro susceptibility testing of zoliflodacin during phase 3 human clinical trials and surveillance studies, and eventually it will be implemented in clinical labs. In a separate study, broth microdilution MIC quality control ranges for zoliflodacin against additional QC strains were determined to be 0.12 to 0.5 µg/ml for Staphylococcus aureus ATCC 29213, 0.25 to 2 µg/ml for Enterococcus faecalis ATCC 29212, 1 to 4 µg/ml for Escherichia coli ATCC 25922, 0.12 to 0.5 µg/ml for Streptococcus pneumoniae ATCC 49619, and 0.12 to 1 µg/ml for Haemophilus influenzae ATCC 49247. These MIC QC ranges were also approved by CLSI for use in future in vitro susceptibility testing studies against organisms other than N. gonorrhoeae.


Asunto(s)
Antibacterianos/farmacología , Barbitúricos/farmacología , Inhibidores Enzimáticos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Pruebas de Sensibilidad Microbiana/métodos , Pruebas de Sensibilidad Microbiana/normas , Compuestos de Espiro/farmacología , Medios de Cultivo , Isoxazoles , Morfolinas , Oxazolidinonas , Control de Calidad
19.
Artículo en Inglés | MEDLINE | ID: mdl-31332073

RESUMEN

Exebacase, a recombinantly produced lysin (cell wall hydrolase), and comparator antibiotics were tested by the broth microdilution method against strain sets of Staphylococcus and Streptococcus spp., which are the most common causes of infective endocarditis in humans. Exebacase was active against all Staphylococcus spp. tested, including S. aureus and coagulase-negative staphylococci (MIC50/90, 0.5/1 µg/ml). Activity against Streptococcus spp. was variable, with S. pyogenes, S. agalactiae, and S. dysgalactiae (MIC50/90, 1/2 µg/ml) among the most susceptible.


Asunto(s)
Antibacterianos/farmacología , Endocarditis Bacteriana/microbiología , Staphylococcus/efectos de los fármacos , Streptococcus/efectos de los fármacos , Endopeptidasas/farmacología , Pruebas de Sensibilidad Microbiana
20.
Nat Rev Microbiol ; 17(7): 459, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31040389

RESUMEN

In figure 1 of the above article, the structure of ETX2514 was missing a double bond and methyl group. This has now been corrected in the PDF and online. The publisher apologizes to the authors and to the readers for this error.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...