Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Antimicrob Agents Chemother ; 67(10): e0048023, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37695298

RESUMEN

A double ampC (AmpCG183D) and ampD (AmpDH157Y) genes mutations have been identified by whole genome sequencing in a Pseudomonas aeruginosa (PaS) that became resistant (PaR) in a patient treated by ceftolozane/tazobactam (C/T). To precisely characterize the respective contributions of these mutations on the decreased susceptibility to C/T and on the parallel increased susceptibility to imipenem (IMI), mutants were generated by homologous recombination in PAO1 reference strain (PAO1- AmpCG183D, PAO1-AmpDH157Y, PAO1-AmpCG183D/AmpDH157Y) and in PaR (PaR-AmpCPaS/AmpDPaS). Sequential time-kill curve experiments were conducted on all strains and analyzed by semi-mechanistic PKPD modeling. A PKPD model with adaptation successfully described the data, allowing discrimination between initial and time-related (adaptive resistance) effects of mutations. With PAO1 and mutant-derived strains, initial EC50 values increased by 1.4, 4.1, and 29-fold after AmpCG183D , AmpDH157Y and AmpCG183D/AmpDH157Y mutations, respectively. EC50 values were increased by 320, 12.4, and 55-fold at the end of the 2 nd experiment. EC50 of PAO1-AmpCG183D/AmpDH157Y was higher than that of single mutants at any time of the experiments. Within the PaR clinical background, reversal of AmpCG183D, and AmpDH157Y mutations led to an important decrease of EC50 value, from 80.5 mg/L to 6.77 mg/L for PaR and PaR-AmpCPaS/AmpDPaS, respectively. The effect of mutations on IMI susceptibility mainly showed that the AmpCG183D mutation prevented the emergence of adaptive resistance. The model successfully described the separate and combined effect of AmpCG183D and AmpDH157Y mutations against C/T and IMI, allowing discrimination and quantification of the initial and time-related effects of mutations. This method could be reproduced in clinical strains to decipher complex resistance mechanisms.


Asunto(s)
Farmacorresistencia Bacteriana , Pseudomonas aeruginosa , Humanos , Antibacterianos/farmacología , Proteínas Bacterianas/genética , beta-Lactamasas/farmacología , Cefalosporinas/farmacología , Imipenem/farmacología , Pruebas de Sensibilidad Microbiana , Mutación , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética , Infecciones por Pseudomonas/tratamiento farmacológico , Tazobactam/farmacología , Farmacorresistencia Bacteriana/genética
2.
Antimicrob Agents Chemother ; 67(7): e0047523, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37310216

RESUMEN

Infections caused by extensively drug-resistant Pseudomonas aeruginosa are difficult to treat due to limited effective treatment options. In this issue, a patient with a corneal infection caused by a Verona integron-encoded metallo-ß-lactamase (VIM)- and Guiana extended-spectrum ß-lactamase (GES)-coproducing P. aeruginosa strain associated with the recent artificial tears-related outbreak in the United States is described. This resistance genotype/phenotype further compromises therapeutic options, and this report provides insights into diagnostic and treatment approaches for clinicians dealing with infections due to this highly resistant P. aeruginosa.


Asunto(s)
Infecciones por Pseudomonas , Pseudomonas aeruginosa , Humanos , Pseudomonas aeruginosa/genética , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/epidemiología , beta-Lactamasas/genética , beta-Lactamasas/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/farmacología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Pruebas de Sensibilidad Microbiana
3.
Biomed Pharmacother ; 164: 114949, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37267640

RESUMEN

Multidrug-resistant bacteria, such as ESBL producing-Klebsiella pneumoniae, have increased substantially, encouraging the development of complementary therapies such as photodynamic inactivation (PDI). PDI uses photosensitizer (PS) compounds that kill bacteria using light to produce reactive oxygen species. We test Ru-based PS to inhibit K. pneumoniae and advance in the characterization of the mode of action. The PDI activity of PSRu-L2, and PSRu-L3, was determined by serial micro dilutions exposing K. pneumoniae to 0.612 J/cm 2 of light dose. PS interaction with cefotaxime was determined on a collection of 118 clinical isolates of K. pneumoniae. To characterize the mode of action of PDI, the bacterial response to oxidative stress was measured by RT-qPCR. Also, the cytotoxicity on mammalian cells was assessed by trypan blue exclusion. Over clinical isolates, the compounds are bactericidal, at doses of 8 µg/mL PSRu-L2 and 4 µg/mL PSRu-L3, inhibit bacterial growth by 3 log10 (>99.9%) with a lethality of 30 min. A remarkable synergistic effect of the PSRu-L2 and PSRu-L3 compounds with cefotaxime increased the bactericidal effect in a subpopulation of 66 ESBL-clinical isolates to > 6 log10 with an FIC-value of 0.16 and 0.17, respectively. The bacterial transcription response suggests that the mode of action occurs through Type II oxidative stress. The upregulation of the extracytoplasmic virulence factors mrkD, magA, and rmpA accompanied this response. Also, the compounds show little or no toxicity in vitro on HEp-2 and HEK293T cells. Through the type II effect, PSs compounds are bactericidal, synergistic on K. pneumoniae, and have low cytotoxicity in mammals.


Asunto(s)
Cefotaxima , Fotoquimioterapia , Animales , Humanos , Cefotaxima/farmacología , Klebsiella pneumoniae , Células HEK293 , beta-Lactamasas/farmacología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Pruebas de Sensibilidad Microbiana , Mamíferos
4.
Ann Transplant ; 28: e939258, 2023 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-37190675

RESUMEN

BACKGROUND Carbapenems are the primary treatment for urinary tract infections (UTIs) caused by extended-spectrum beta-lactamase (ESBL)-producing Klebsiella pneumoniae. However, the recurrence rate is high, and patients often require rehospitalization. We present the results of an observational study on patients with recurrent UTIs who were treated in an outpatient setting with maximal therapeutic oral doses of amoxicillin with clavulanic acid. MATERIAL AND METHODS All patients had pyuria and ESBL-producing K. pneumoniae in urine culture. The starting dosage was 2875 g of amoxicillin twice daily and 125 mg of clavulanic acid twice daily. We down-titrated the doses every 7-14 days and continued prophylactic therapy with amoxicillin/clavulanic acid at 250/125 mg for up to 3 months. We defined therapeutic failure as ESBL-positive K. pneumoniae in urine culture during therapy and recurrence as positive urine culture with the same strain within 1 month after the end of treatment. RESULTS We included 9 patients: 7 kidney graft recipients, 1 liver graft recipient, and 1 patient with chronic kidney disease. We observed no therapeutic failures and no recurrences in the study group during the study period. In 1 case, the patient experienced a subsequent UTI caused by ESBL-producing K. pneumoniae 4 months after completing the therapy. CONCLUSIONS In conclusion, it is possible to break the resistance of ESBL-producing K. pneumoniae strains with high doses of oral amoxicillin with clavulanic acid. Such treatment could be an alternative to carbapenems in select cases.


Asunto(s)
Infecciones por Klebsiella , Infecciones Urinarias , Humanos , Klebsiella pneumoniae , Antibacterianos/uso terapéutico , Amoxicilina/uso terapéutico , Amoxicilina/farmacología , Ácido Clavulánico/uso terapéutico , Ácido Clavulánico/farmacología , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/etiología , Pruebas de Sensibilidad Microbiana , Farmacorresistencia Bacteriana , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/etiología , Carbapenémicos/farmacología , Carbapenémicos/uso terapéutico , beta-Lactamasas/farmacología , beta-Lactamasas/uso terapéutico
5.
J Antimicrob Chemother ; 78(5): 1211-1218, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-36897329

RESUMEN

BACKGROUND: While the treatment of ESBL-producing Enterobacterales osteomyelitis relies on carbapenems, the optimal regimen for OXA48 types remains unclear. We evaluated the efficacy of ceftazidime/avibactam in different combinations in an experimental model of OXA-48-/ESBL-producing Escherichia coli osteomyelitis. METHODS: E. coli pACYC184 is a clinical strain harbouring blaOXA-48 and blaCTX-M-15 inserts, with 'increased exposure susceptibility' to imipenem (MIC, 2 mg/L), gentamicin (MIC, 0.5 mg/L), colistin (MIC, 0.25 mg/L), ceftazidime/avibactam (MIC, 0.094 mg/L) and fosfomycin (MIC, 1 mg/L), and resistance to ceftazidime (MIC, 16 mg/L). Osteomyelitis was induced in rabbits by tibial injection of 2 × 108 cfu of OXA-48/ESBL E. coli. Treatment started 14 days later for 7 days in six groups: (1) control, (2) colistin 150.000 IU/kg subcutaneously (SC) q8h, (3) ceftazidime/avibactam 100/25 mg/kg SC q8h, (4) ceftazidime/avibactam + colistin, (5) ceftazidime/avibactam + fosfomycin 150 mg/kg SC q12h, (6) ceftazidime/avibactam + gentamicin 15 mg/kg intramuscularly (IM) q24h. Treatment was evaluated at Day 24 according to bone cultures. RESULTS: In vitro, time-kill curves of ceftazidime/avibactam in combination showed a synergistic effect. In vivo, compared with controls, rabbits treated with colistin alone had similar bone bacterial density (P = 0.50), whereas ceftazidime/avibactam alone or in combinations significantly decreased bone bacterial densities (P = 0.004 and P < 0.0002, respectively). Bone sterilization was achieved using ceftazidime/avibactam in combination with colistin (91%) or fosfomycin (100%) or gentamicin (100%) (P < 0.0001), whereas single therapies were not different from controls. No ceftazidime/avibactam-resistant strains emerged in rabbits treated, regardless of the combination. CONCLUSIONS: In our model of E. coli OXA-48/ESBL osteomyelitis, ceftazidime/avibactam in combination was more effective than any single therapy, whatever the companion drug used (gentamicin or colistin or fosfomycin).


Asunto(s)
Fosfomicina , Osteomielitis , Animales , Conejos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Escherichia coli , Fosfomicina/uso terapéutico , Fosfomicina/farmacología , Colistina/farmacología , beta-Lactamasas/farmacología , Compuestos de Azabiciclo/farmacología , Combinación de Medicamentos , Gentamicinas/farmacología , Osteomielitis/tratamiento farmacológico , Pruebas de Sensibilidad Microbiana
6.
Biol Pharm Bull ; 46(3): 394-398, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36543225

RESUMEN

Carbapenem-resistant bacterial infections pose an urgent threat to public health worldwide. Horizontal transmission of the ß-lacatamase Klebsiella pneumoniae carbapenemase (blaKPC) multidrug resistance gene is a major mechanism for global dissemination of carbapenem resistance. Here, we investigated the effects of baicalein, an active ingredient of a Chinese herbal medicine, on plasmid-mediated horizontal transmission of blaKPC from a meropenem-resistant K. pneumoniae strain (JZ2157) to a meropenem-sensitive Escherichia coli strain (E600). Baicalein showed no direct effects on the growth of JZ2157 or E600. Co-cultivation of JZ2157 and E600 caused the spread of meropenem resistance from JZ2157 to E600. Baicalein at 40 and 400 µg/mL significantly inhibited the spread of meropenem resistance. Co-cultivation also resulted in plasmid-mediated transmission of blaKPC from JZ2157 to E600, which was inhibited by baicalein. Therefore, baicalein may be used in clinical practice to prevent or contain outbreaks of carbapenem-resistant infections by inhibiting the horizontal transfer of resistance genes across bacteria species.


Asunto(s)
Infecciones por Klebsiella , Klebsiella pneumoniae , Humanos , Klebsiella pneumoniae/genética , Escherichia coli , Meropenem/farmacología , Genes MDR , Paraoxon/farmacología , beta-Lactamasas/genética , beta-Lactamasas/farmacología , Proteínas Bacterianas/genética , Plásmidos , Carbapenémicos/farmacología , Antibacterianos/farmacología , Pruebas de Sensibilidad Microbiana
7.
Food Funct ; 13(19): 9761-9771, 2022 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-36124641

RESUMEN

The antimicrobial actions of natural compounds derived from medicinal plants have been well documented. However, their detailed mechanisms underlying the action against microorganisms remain largely unexplored. Salmonella enterica is a common pathogen causing both gastrointestinal and systemic diseases. In Salmonella enterica, the type III secretion system (T3SS) is employed to export secreted effectors directly to the cytoplasm of host cells. Using a SipA-ß-lactamase reporter, we found that hyperoside (HYP) inhibited the activity of Salmonella T3SS needle protein InvG, prevented damage to host cells and protected mice against Salmonella enterica serovar Typhimurium. It was also observed that HYP binds to InvG directly through hydrogen-bridged cations and hydrophobic interactions. The unique mechanism of antibacterial action of HYP suggested that it could be used as a potentially effective candidate for future antimicrobial regimens.


Asunto(s)
Salmonella enterica , Salmonella typhimurium , Animales , Antibacterianos/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cationes , Hidrógeno/farmacología , Ratones , Quercetina/análogos & derivados , Sistemas de Secreción Tipo III/metabolismo , beta-Lactamasas/metabolismo , beta-Lactamasas/farmacología
8.
Drugs ; 82(5): 533-557, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35294769

RESUMEN

Sulopenem (formerly known as CP-70,429, and CP-65,207 when a component of a racemic mixture with its R isomer) is an intravenous and oral penem that possesses in vitro activity against fluoroquinolone-resistant, extended spectrum ß-lactamases (ESBL)-producing, multidrug-resistant (MDR) Enterobacterales. Sulopenem is being developed to treat patients with uncomplicated and complicated urinary tract infections (UTIs) as well as intra-abdominal infections. This review will focus mainly on its use in UTIs. The chemical structure of sulopenem shares properties of penicillins, cephalosporins, and carbapenems. Sulopenem is available as an oral prodrug formulation, sulopenem etzadroxil, which is hydrolyzed by intestinal esterases, resulting in active sulopenem. In early studies, the S isomer of CP-65,207, later developed as sulopenem, demonstrated greater absorption, higher drug concentrations in the urine, and increased stability against the renal enzyme dehydropeptidase-1 compared with the R isomer, which set the stage for its further development as a UTI antimicrobial. Sulopenem is active against both Gram-negative and Gram-positive microorganisms. Sulopenem's ß-lactam ring alkylates the serine residues of penicillin-binding protein (PBP), which inhibits peptidoglycan cross-linking. Due to its ionization and low molecular weight, sulopenem passes through outer membrane proteins to reach PBPs of Gram-negative bacteria. While sulopenem activity is unaffected by many ß-lactamases, resistance arises from alterations in PBPs (e.g., methicillin-resistant Staphylococcus aureus [MRSA]), expression of carbapenemases (e.g., carbapenemase-producing Enterobacterales and in Stenotrophomonas maltophilia), reduction in the expression of outer membrane proteins (e.g., some Klebsiella spp.), and the presence of efflux pumps (e.g., MexAB-OprM in Pseudomonas aeruginosa), or a combination of these mechanisms. In vitro studies have reported that sulopenem demonstrates greater activity than meropenem and ertapenem against Enterococcus faecalis, Listeria monocytogenes, methicillin-susceptible S. aureus (MSSA), and Staphylococcus epidermidis, as well as similar activity to carbapenems against Streptococcus agalactiae, Streptococcus pneumoniae, and Streptococcus pyogenes. With some exceptions, sulopenem activity against Gram-negative aerobes was less than ertapenem and meropenem but greater than imipenem. Sulopenem activity against Escherichia coli carrying ESBL, CTX-M, or Amp-C enzymes, or demonstrating MDR phenotypes, as well as against ESBL-producing Klebsiella pneumoniae, was nearly identical to ertapenem and meropenem and greater than imipenem. Sulopenem exhibited identical or slightly greater activity than imipenem against many Gram-positive and Gram-negative anaerobes, including Bacteroides fragilis. The pharmacokinetics of intravenous sulopenem appear similar to carbapenems such as imipenem-cilastatin, meropenem, and doripenem. In healthy subjects, reported volumes of distribution (Vd) ranged from 15.8 to 27.6 L, total drug clearances (CLT) of 18.9-24.9 L/h, protein binding of approximately 10%, and elimination half-lives (t½) of 0.88-1.03 h. The estimated renal clearance (CLR) of sulopenem is 8.0-10.6 L/h, with 35.5% ± 6.7% of a 1000 mg dose recovered unchanged in the urine. An ester prodrug, sulopenem etzadroxil, has been developed for oral administration. Initial investigations reported a variable oral bioavailability of 20-34% under fasted conditions, however subsequent work showed that bioavailability is significantly improved by administering sulopenem with food to increase its oral absorption or with probenecid to reduce its renal tubular secretion. Food consumption increases the area under the curve (AUC) of oral sulopenem (500 mg twice daily) by 23.6% when administered alone and 62% when administered with 500 mg of probenecid. Like carbapenems, sulopenem demonstrates bactericidal activity that is associated with the percentage of time that free concentrations exceed the MIC (%f T > MIC). In animal models, bacteriostasis was associated with %f T > MICs ranging from 8.6 to 17%, whereas 2-log10 kill was seen at values ranging from 12 to 28%. No pharmacodynamic targets have been documented for suppression of resistance. Sulopenem concentrations in urine are variable, ranging from 21.8 to 420.0 mg/L (median 84.4 mg/L) in fasted subjects and 28.8 to 609.0 mg/L (median 87.3 mg/L) in those who were fed. Sulopenem has been compared with carbapenems and cephalosporins in guinea pig and murine systemic and lung infection animal models. Studied pathogens included Acinetobacter calcoaceticus, B. fragilis, Citrobacter freundii, Enterobacter cloacae, E. coli, K. pneumoniae, Proteus vulgaris, and Serratia marcescens. These studies reported that overall, sulopenem was non-inferior to carbapenems but appeared to be superior to cephalosporins. A phase III clinical trial (SURE-1) reported that sulopenem was not non-inferior to ciprofloxacin in women infected with fluoroquinolone-susceptible pathogens, due to a higher rate of asymptomatic bacteriuria in sulopenem-treated patients at the test-of-cure visit. However, the researchers reported superiority of sulopenem etzadroxil/probenecid over ciprofloxacin for the treatment of uncomplicated UTIs in women infected with fluoroquinolone/non-susceptible pathogens, and non-inferiority in all patients with a positive urine culture. A phase III clinical trial (SURE-2) compared intravenous sulopenem followed by oral sulopenem etzadroxil/probenecid with ertapenem in the treatment of complicated UTIs. No difference in overall success was noted at the end of therapy. However, intravenous sulopenem followed by oral sulopenem etzadroxil was not non-inferior to ertapenem followed by oral stepdown therapy in overall success at test-of-cure due to a higher rate of asymptomatic bacteriuria in the sulopenem arm. After a meeting with the US FDA, Iterum stated that they are currently evaluating the optimal design for an additional phase III uncomplicated UTI study to be conducted prior to the potential resubmission of the New Drug Application (NDA). It is unclear at this time whether Iterum intends to apply for EMA or Japanese regulatory approval. The safety and tolerability of sulopenem has been reported in various phase I pharmacokinetic studies and phase III clinical trials. Sulopenem (intravenous and oral) appears to be well tolerated in healthy subjects, with and without the coadministration of probenecid, with few serious drug-related treatment-emergent adverse events (TEAEs) reported to date. Reported TEAEs affecting ≥1% of patients were (from most to least common) diarrhea, nausea, headache, vomiting and dizziness. Discontinuation rates were low and were not different than comparator agents. Sulopenem administered orally and/or intravenously represents a potentially well tolerated and effective option for treating uncomplicated and complicated UTIs, especially in patients with documented or highly suspected antimicrobial pathogens to commonly used agents (e.g. fluoroquinolone-resistant E. coli), and in patients with documented microbiological or clinical failure or patients who demonstrate intolerance/adverse effects to first-line agents. This agent will likely be used orally in the outpatient setting, and intravenously followed by oral stepdown in the hospital setting. Sulopenem also allows for oral stepdown therapy in the hospital setting from intravenous non-sulopenem therapy. More clinical data are required to fully assess the clinical efficacy and safety of sulopenem, especially in patients with complicated UTIs caused by resistant pathogens such as ESBL-producing, Amp-C, MDR E. coli. Antimicrobial stewardship programs will need to create guidelines for when this oral and intravenous penem should be used.


Asunto(s)
Bacteriuria , Staphylococcus aureus Resistente a Meticilina , Profármacos , Infecciones Urinarias , Animales , Femenino , Cobayas , Humanos , Masculino , Ratones , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bacteriuria/inducido químicamente , Bacteriuria/tratamiento farmacológico , beta-Lactamasas/farmacología , Carbapenémicos/farmacología , Cefalosporinas/farmacología , Ciprofloxacina/farmacología , Ertapenem , Escherichia coli , Fluoroquinolonas/farmacología , Bacterias Gramnegativas , Imipenem/farmacología , Lactamas , Proteínas de la Membrana/farmacología , Meropenem/farmacología , Probenecid/farmacología , Profármacos/farmacología , Staphylococcus aureus , Infecciones Urinarias/tratamiento farmacológico
9.
J Antibiot (Tokyo) ; 75(5): 282-286, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35197545

RESUMEN

The emergence of bacterial resistance poses a serious threat to public health. One of the most important resistance mechanisms against ß-lactam antibiotics is the production of metallo-ß-lactamases (MBLs). In this study, α-lipoic acid (LA) and methimazole (MMI), which have been used in clinical practice as non-antibacterial drugs and as a supplement, were chosen to explore their potential to be metallo-ß-lactamases inhibitors (MBLIs). Enzyme inhibition assays showed that LA and MMI had moderate inhibitory activity against NDM-1 but no activity against VIM-2 and IMP-7. Antibacterial assays to determine synergy, demonstrated that the combination of LA or MMI with meropenem (MER) reduced the MIC value of MER against NDM-1 producing E. coli 16 times and 4 times, respectively, lower than that of MER alone. The fractional inhibitory concentration index (FICI) values were calculated to be less than 0.5, indicating that both LA and MMI had synergistic antibacterial effects with MER against all three MBLs expressing E. coli strains. The time-kill studies also suggested that LA and MMI were effective in restoring the antibacterial effect of MER. These findings revealed that LA and MMI are potential carbapenem enhancers, and provide a starting point for the development of potent MBLIs.


Asunto(s)
Ácido Tióctico , Inhibidores de beta-Lactamasas , Antibacterianos/farmacología , Escherichia coli , Meropenem/farmacología , Metimazol/farmacología , Pruebas de Sensibilidad Microbiana , Ácido Tióctico/farmacología , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/farmacología
10.
Eur J Hosp Pharm ; 29(2): 113-115, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34716170

RESUMEN

The case of a female who had an accident that caused an open fracture is reported. During hospitalisation, Verona integron-encoded metallo-ß-lactamase (VIM)-producing Klebsiella pneumoniae was isolated. Antimicrobial susceptibility testing revealed resistance to ß-lactam antibiotics, quinolones, trimethoprim/sulfamethoxazole, and susceptibility to tigecycline, colistin, fosfomycin and aminoglycosides. Synergistic activity of ceftazidime-avibactam and aztreonam was proved in vitro and a combined therapy with tigecycline was started. Combination with aminoglycosides was ruled out as it was not described in the literature and also in order to avoid side effects. Colistin was rejected because of its nephrotoxicity profile. The antibiotic treatment was assessed by a multidisciplinary team with a pharmacist who closely monitored adverse effects and interactions with other drugs. The total duration of this combination was 25 days, without any adverse events reported. Fourteen weeks after the accident the patient was discharged. After 2 months of follow-up neither relapses nor reinfections have been reported.


Asunto(s)
Aztreonam , Ceftazidima , Compuestos de Azabiciclo , Aztreonam/farmacología , Aztreonam/uso terapéutico , Ceftazidima/farmacología , Ceftazidima/uso terapéutico , Combinación de Medicamentos , Femenino , Humanos , Integrones , Klebsiella pneumoniae/metabolismo , Pruebas de Sensibilidad Microbiana , beta-Lactamasas/metabolismo , beta-Lactamasas/farmacología
11.
Int J Antimicrob Agents ; 58(1): 106361, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34000372

RESUMEN

OBJECTIVES: To compare the efficacy of temocillin with carbapenems for extended spectrum ß-lactamase (ESBL)-producing Enterobacteriaceae urinary tract infections (ESBL-E UTI). METHODS: A multicenter retrospective case-control study of adults with ESBL-E UTI was conducted between January 2015 and October 2019. Cases received temocillin ≥ 50% of the effective antibiotic therapy duration and controls exclusively received carbapenem; they were statistically matched (1:1 ratio) on 6-month period, sex and age. The clinical cure at the end of antibiotic therapy was analysed using conditional logistic regression. RESULTS: Seventy-two temocillin cases were matched to 72 carbapenem controls. Most (67%) were male, median age was 69.4 years, 81 (56%) were immunocompromised, including 44 (31%) solid organ transplant recipients. There was no difference between cases and controls for baseline characteristics and microorganisms involved: Klebsiella pneumoniae in 59 (41%), Escherichia coli in 57 (40%), and Enterobacter spp. in 24 (17%). The median time from admission to effective antibiotic therapy was 0 days [range, 0-2]. Among cases, first-line antibiotic therapy (≤ 72 hours) was temocillin in six (8%) and carbapenems in 39 (54%). Temocillin was given at the median daily dose of 4 g [range, 2-4] after 3 days [range, 2-5] of carbapenems. Patients received temocillin for 81% [range, 70-93] of the effective antibiotic course duration over 11 days [range, 8-14]. The effective antibiotic duration was similar in cases and controls (P = 0.067). Clinical cure at the end of antibiotic therapy was 94% (68/72) in cases vs. 99% (71/72) in controls (P = 0.206), with no difference among immunocompromised and solid organ transplant patients (P > 0.050). CONCLUSIONS: Temocillin effectively relayed ß-lactams, including carbapenems, to treat (complicated) ESBL-E UTI. Its efficacy was consistent among kidney transplant recipients.


Asunto(s)
Carbapenémicos/farmacología , Infecciones por Enterobacteriaceae/dietoterapia , Infecciones por Enterobacteriaceae/microbiología , Enterobacteriaceae/efectos de los fármacos , Penicilinas/uso terapéutico , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología , Anciano , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana , Femenino , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Estudios Retrospectivos , Resultado del Tratamiento , beta-Lactamasas/metabolismo , beta-Lactamasas/farmacología
12.
Diagn Microbiol Infect Dis ; 98(1): 115080, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32619895

RESUMEN

The objective of this study was to utilize a co-culture hollow-fiber infection model (HFIM) to characterize the interplay between a small, difficult-to-detect, New Delhi metallo-ß-lactamase-producing Klebsiella pneumoniae (NDM-Kp) minor population and a larger K. pneumoniae carbapenemase (KPC)-producing K. pneumoniae population in the presence of KPC-directed antibacterial therapy. Selective plating onto agar with ceftazidime-avibactam was used to track the density of the NDM-Kp population. Susceptibility testing and the Verigene System failed to identify the small initial NDM-Kp population. However, a ceftazidime-avibactam Etest detected resistant colonies that were confirmed to be NDM-Kp. In the HFIM, all of the investigated drug regimens caused regrowth within 24 h and resulted in >109 CFU/mL of NDM-Kp. Our study demonstrates that the HFIM is a powerful tool for studying the population dynamics of multiple pathogens during antimicrobial exposure and also highlights that difficult-to-detect minor populations of drug-resistant bacteria may cause treatment failure without appropriate antibacterial therapy.


Asunto(s)
Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/crecimiento & desarrollo , Pruebas de Sensibilidad Microbiana/métodos , beta-Lactamasas/farmacología , Compuestos de Azabiciclo/farmacología , Proteínas Bacterianas/farmacología , Ceftazidima/farmacología , Combinación de Medicamentos , Humanos , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/microbiología , Viabilidad Microbiana
13.
Antimicrob Agents Chemother ; 64(10)2020 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-32718961

RESUMEN

This study aimed to evaluate the antimicrobial activity of the novel monosulfactam 0073 against multidrug-resistant Gram-negative bacteria in vitro and in vivo and to characterize the mechanisms underlying 0073 activity. The in vitro activities of 0073, aztreonam, and the combination with avibactam were assessed by MIC and time-kill assays. The safety of 0073 was evaluated using 3-(4,5-dimethylthizol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and acute toxicity assays. Murine thigh infection and pneumonia models were employed to define in vivo efficacy. A penicillin-binding protein (PBP) competition assay and confocal microscopy were conducted. The inhibitory action of 0073 against ß-lactamases was evaluated by the half-maximal inhibitory concentration (IC50), and resistance development was evaluated via serial passage. The monosulfactam 0073 showed promising antimicrobial activity against Enterobacteriaceae, Pseudomonas aeruginosa, and Acinetobacter baumannii isolates producing metallo-ß-lactamases (MBLs) and serine ß-lactamases. In preliminary experiments, compound 0073 exhibited safety both in vitro and in vivo In the murine thigh infection model and the pneumonia models in which infection was induced by P. aeruginosa and Klebsiella pneumoniae, 0073 significantly reduced the bacterial burden. Compound 0073 targeted several PBPs and exerted inhibitory effects against some serine ß-lactamases. Finally, 0073 showed a reduced propensity for resistance selection compared with that of aztreonam. The novel monosulfactam 0073 exhibited increased activity against ß-lactamase-producing Gram-negative organisms compared with the activity of aztreonam and showed good safety profiles both in vitro and in vivo The underlying mechanisms may be attributed to the affinity of 0073 for several PBPs and its inhibitory activity against some serine ß-lactamases. These data indicate that 0073 represents a potential treatment for infections caused by ß-lactamase-producing multidrug-resistant bacteria.


Asunto(s)
Antibacterianos , Compuestos de Azabiciclo , beta-Lactamasas/farmacología , Animales , Antibacterianos/farmacología , Aztreonam , Enterobacteriaceae , Ratones , Pruebas de Sensibilidad Microbiana , Inhibidores de beta-Lactamasas
14.
Enferm. infecc. microbiol. clín. (Ed. impr.) ; 35(8): 499-504, oct. 2017. graf, tab
Artículo en Inglés | IBECS | ID: ibc-167837

RESUMEN

Introduction: Antimicrobial resistance in Enterobacteriaceae is increasing worldwide and is making treating infections caused by multidrug-resistant Enterobacteriaceae a challenge. The use of Beta -lactam agents is compromised by microorganisms harboring extended-spectrum Beta -lactamases (ESBLs) and other mechanisms of resistance. Avibactam is a non Beta -lactam agent that inhibits clinically relevant Beta -lactamases, such as ESBL and AmpC. The ceftazidime-avibactam combination (CAZ-AVI) was recently approved for use in certain complicated infections, and may provide a therapeutic alternative for infections caused by these microorganisms. Methods: The in vitro activity of CAZ and CAZ-AVI (AVI at a fixed concentration of 4mg/L) was tested against 250 clinical isolates of Enterobacteriaceae using broth microdilution. EUCAST breakpoint criteria were used for CAZ, and FDA criteria for CAZ-AVI. Clinical isolates included bacteria producing extended-spectrum Beta -lactamases (ESBLs) and acquired AmpC Beta -lactamases (AACBLs). Porin loss in Klebsiella pneumoniae was also evaluated. Results: The combination of AVI with CAZ displayed excellent activity against clinical isolates of ESBL-producing Escherichia coli and Klebsiella pneumoniae, rendering all the ceftazidime-resistant isolates susceptible to ceftazidime. CAZ-AVI retained activity against porin-deficient isolates of K. pneumoniae producing ESBLs, AACBLs, or both, although MIC values were higher compared to porin-expressing isolates. CAZ-AVI rendered all the ceftazidime-resistant AACBL-producing Enterobacteriaceae tested susceptible to ceftazidime. Conclusion: CAZ-AVI showed potent in vitro activity against clinical isolates of Enterobacteriaceaeproducing ESBLs and/or AACBLs, including K. pneumoniae with loss of porins (AU)


Introducción: La resistencia antibiótica en enterobacterias está en aumento y el tratamiento de infecciones producidas por enterobacterias multirresistentes supone un reto terapéutico. El uso de betalactámicos se afecta con la producción de betalactamasas de espectro extendido (BLEE) y otros mecanismos de resistencia. Avibactam es un compuesto no betalactámico que inhibe betalactamasas como BLEE o AmpC. La combinación ceftazidima-avibactam (CAZ-AVI) ha sido aprobada recientemente para el tratamiento de infecciones complicadas y puede ser una alternativa terapéutica en estas infecciones. Métodos: La actividad in vitro de CAZ y CAZ-AVI (AVI, concentración fija de 4mg/mL) fue determinada en 250 aislamientos clínicos de enterobacterias mediante microdilución en caldo. Los puntos de corte de EUCAST fueron utilizados para CAZ, y los criterios de FDA se utilizaron para CAZ-AVI. Las enterobacterias estudiadas producían BLEE y/o AmpC adquiridas (BLAA). El papel de la pérdida de porinas en Klebsiella pneumoniae también fue evaluado. Resultados: CAZ-AVI demostró una excelente actividad en Escherichia coli y Klebsiella pneumoniaeproductoras de BLEE, devolviendo la sensibilidad a CAZ en todos los aislamientos resistentes a CAZ. CAZ-AVI mantuvo su actividad en aislamientos de K. pneumoniae deficientes en porinas productoras de BLEE y/o BLAA, aunque los valores de CMI fueron más altos comparados con las cepas que expresaban porinas. En todas las enterobacterias resistentes a ceftazidima productoras de BLAA analizadas en este estudio CAZ-AVI devolvió la sensibilidad a ceftazidima. Conclusión: CAZ-AVI demostró una potente actividad in vitro en aislamientos clínicos de enterobacterias productoras de BLEE y/o BLAA, incluyendo K. pneumoniae con pérdida de porinas (AU)


Asunto(s)
Resistencia a Múltiples Medicamentos , Enterobacteriaceae , beta-Lactamasas/farmacología , Inhibidores de beta-Lactamasas/uso terapéutico , Técnicas In Vitro/métodos , Porinas/aislamiento & purificación , Klebsiella pneumoniae , Klebsiella pneumoniae/aislamiento & purificación , Pruebas de Sensibilidad Microbiana/instrumentación
15.
Artículo en Inglés | MEDLINE | ID: mdl-28348155

RESUMEN

The correlation of the clinical efficacy of ceftazidime-avibactam (plus metronidazole) with that of meropenem was evaluated in subjects infected with Gram-negative isolates having characterized ß-lactam resistance mechanisms from the complicated intra-abdominal infection (cIAI) phase 3 clinical trials. Enterobacteriaceae isolates displaying ceftriaxone and/or ceftazidime MIC values of ≥2 µg/ml and Pseudomonas aeruginosa isolates with ceftazidime MIC values of ≥16 µg/ml were characterized for extended-spectrum-ß-lactamase (ESBL) content. Enterobacteriaceae and P. aeruginosa isolates with imipenem and meropenem MIC values of ≥2 and ≥8 µg/ml, respectively, were tested for carbapenemase genes. The primary efficacy endpoint was clinical cure at test of cure (TOC) among the members of the microbiologically modified intention-to-treat (mMITT) population. A total of 14.5% (56/387) and 18.8% (74/394) of patients in the ceftazidime-avibactam and meropenem arms had isolates that met the MIC screening criteria at the baseline visit, respectively. CTX-M variants alone (29.7%; 41/138) or in combination with OXA-1/30 (35.5%; 49/138), most commonly, blaCTX-M group 1 variants (79/90; 87.8%), represented the ß-lactamases most frequently observed among Enterobacteriaceae isolates. Among the patients infected with pathogens that did not meet the screening criteria, 82.2% showed clinical cure in the ceftazidime-avibactam group versus 85.9% in the meropenem group. Among patients infected with any pathogens that met the MIC screening criteria, clinical cure rates at TOC were 87.5% and 86.5% for the ceftazidime-avibactam and meropenem groups, respectively. Ceftazidime-avibactam had clinical cure rates of 92.5% to 90.5% among patients infected with ESBL- and/or carbapenemase-producing Enterobacteriaceae strains at the baseline visit, while meropenem showed rates of 84.9% to 85.4%. The ceftazidime-avibactam and meropenem groups had cure rates of 75.0% and 86.7%, respectively, among patients having any pathogens producing AmpC enzymes. The efficacy of ceftazidime-avibactam was similar to that of meropenem for treatment of cIAI caused by ESBL-producing organisms. (This study has been registered at ClinicalTrials.gov under registration no. NCT01499290 and NCT01500239.).


Asunto(s)
Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bacterias Aerobias Gramnegativas/efectos de los fármacos , Bacterias Aerobias Gramnegativas/patogenicidad , Infecciones Intraabdominales/tratamiento farmacológico , beta-Lactamasas/farmacología , beta-Lactamasas/uso terapéutico , Compuestos de Azabiciclo , Ceftazidima , Combinación de Medicamentos , Escherichia coli/efectos de los fármacos , Escherichia coli/patogenicidad , Humanos , Infecciones Intraabdominales/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/patogenicidad , Pruebas de Sensibilidad Microbiana
16.
Postgrad Med ; 129(2): 242-258, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27712137

RESUMEN

Urinary tract infections (UTIs) caused by antibiotic-resistant Gram-negative bacteria are a growing concern due to limited therapeutic options. Gram-negative bacteria, specifically Enterobacteriaceae, are common causes of both community-acquired and hospital acquired UTIs. These organisms can acquire genes that encode for multiple antibiotic resistance mechanisms, including extended-spectrum-lactamases (ESBLs), AmpC- ß -lactamase, and carbapenemases. The assessment of suspected UTI includes identification of characteristic symptoms or signs, urinalysis, dipstick or microscopic tests, and urine culture if indicated. UTIs are categorized according to location (upper versus lower urinary tract) and severity (uncomplicated versus complicated). Increasing rates of antibiotic resistance necessitate judicious use of antibiotics through the application of antimicrobial stewardship principles. Knowledge of the common causative pathogens of UTIs including local susceptibility patterns are essential in determining appropriate empiric therapy. The recommended first-line empiric therapies for acute uncomplicated bacterial cystitis in otherwise healthy adult nonpregnant females is a 5-day course of nitrofurantion or a 3-g single dose of fosfomycin tromethamine. Second-line options include fluoroquinolones and ß-lactams, such as amoxicillin-clavulanate. Current treatment options for UTIs due to AmpC- ß -lactamase-producing organisms include fosfomycin, nitrofurantion, fluoroquinolones, cefepime, piperacillin-tazobactam and carbapenems. In addition, treatment options for UTIs due to ESBLs-producing Enterobacteriaceae include nitrofurantion, fosfomycin, fluoroquinolones, cefoxitin, piperacillin-tazobactam, carbapenems, ceftazidime-avibactam, ceftolozane-tazobactam, and aminoglycosides. Based on identification and susceptibility results, alternatives to carbapenems may be used to treat mild-moderate UTIs caused by ESBL-producing Enterobacteriaceae. Ceftazidime-avibactam, colistin, polymixin B, fosfomycin, aztreonam, aminoglycosides, and tigecycline are treatment options for UTIs caused by carbapenem-resistant Enterobacteriaceae (CRE). Treatment options for UTIs caused by multidrug resistant (MDR)-Pseudomonas spp. include fluoroquinolones, ceftazidime, cefepime, piperacillin-tazobactam, carbapenems, aminoglycosides, colistin, ceftazidime-avibactam, and ceftolozane-tazobactam. The use of fluoroquinolones for empiric treatment of UTIs should be restricted due to increased rates of resistance. Aminoglycosides, colistin, and tigecycline are considered alternatives in the setting of MDR Gram-negative infections in patients with limited therapeutic options.


Asunto(s)
Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana/fisiología , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Infecciones Urinarias/tratamiento farmacológico , beta-Lactamasas/farmacología , Aminoglicósidos/farmacología , Aminoglicósidos/uso terapéutico , Proteínas Bacterianas/genética , Proteínas Bacterianas/farmacología , Comorbilidad , Fluoroquinolonas/farmacología , Fluoroquinolonas/uso terapéutico , Fosfomicina/farmacología , Fosfomicina/uso terapéutico , Humanos , Pruebas de Sensibilidad Microbiana , Minociclina/farmacología , Minociclina/uso terapéutico , Nitrofurantoína/farmacología , Nitrofurantoína/uso terapéutico , Índice de Severidad de la Enfermedad , Infecciones Urinarias/diagnóstico , beta-Lactamasas/genética , beta-Lactamas/farmacología , beta-Lactamas/uso terapéutico
17.
Anaerobe ; 41: 58-67, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27262694

RESUMEN

The gut microbiome, composed of the microflora that inhabit the gastrointestinal tract and their genomes, make up a complex ecosystem that can be disrupted by antibiotic use. The ensuing dysbiosis is conducive to the emergence of opportunistic pathogens such as Clostridium difficile. A novel approach to protect the microbiome from antibiotic-mediated dysbiosis is the use of beta-lactamase enzymes to degrade residual antibiotics in the gastrointestinal tract before the microflora are harmed. Here we present the preclinical development and early clinical studies of the beta-lactamase enzymes, P3A, currently referred to as SYN-004, and its precursor, P1A. Both P1A and SYN-004 were designed as orally-delivered, non-systemically available therapeutics for use with intravenous beta-lactam antibiotics. SYN-004 was engineered from P1A, a beta-lactamase isolated from Bacillus licheniformis, to broaden its antibiotic degradation profile. SYN-004 efficiently hydrolyses penicillins and cephalosporins, the most widely used IV beta-lactam antibiotics. In animal studies, SYN-004 degraded ceftriaxone in the GI tract of dogs and protected the microbiome of pigs from ceftriaxone-induced changes. Phase I clinical studies demonstrated SYN-004 safety and tolerability. Phase 2 studies are in progress to assess the utility of SYN-004 for the prevention of antibiotic-associated diarrhea and Clostridium difficile disease.


Asunto(s)
Antibacterianos/farmacología , Enterocolitis Seudomembranosa/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Proteínas Recombinantes/farmacología , beta-Lactamasas/farmacología , Animales , Antibacterianos/química , Antibacterianos/uso terapéutico , Clostridioides difficile/efectos de los fármacos , Perros , Farmacorresistencia Bacteriana , Estabilidad de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Cinética , Pruebas de Sensibilidad Microbiana , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapéutico , Sus scrofa , beta-Lactamasas/química , beta-Lactamasas/uso terapéutico
18.
Medicine (Baltimore) ; 95(7): e2243, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26886594

RESUMEN

Transplant recipients are at high risk of infections caused by multidrug resistant microorganisms. Due to the limited therapeutic options, innovative antimicrobial combinations against carbapenem-resistant Enterobacteriaceae causing severe infections are necessary.A 61-year-old woman with a history of congenital solitary kidney underwent renal transplantation. The postoperative course was complicated by nosocomial pneumonia due to Stenotrophomonas maltophilia and pan-sensitive Escherichia coli, successfully treated with antimicrobial therapy. On postoperative day 22, diagnosis of surgical site infection and nosocomial pneumonia with concomitant bacteremia due to a Klebisella pneumoniae carbapenemase-producer E coli was made. The patient was treated with the double-carbapenem regimen (high dose of meropenem plus ertapenem) and a potent synergistic and bactericidal activity of this un-conventional therapeutic strategy was observed in vitro. Despite a microbiological response with prompt negativity of blood cultures, the patient faced a worse outcome because of severe hemorrhagic shock.The double-carbapenem regimen might be considered as a rescue therapy in those subjects, including transplant recipients, in whom previous antimicrobial combinations failed or when colistin use might be discouraged. Performing in vitro synergy testing should be strongly encouraged in cases of infections caused by pan-drug resistant strains, especially in high-risk patients.


Asunto(s)
Antibacterianos/uso terapéutico , Bacteriemia/tratamiento farmacológico , Infecciones por Escherichia coli/tratamiento farmacológico , Escherichia coli , Trasplante de Riñón , Antibacterianos/farmacología , Bacteriemia/microbiología , Proteínas Bacterianas/farmacología , Carbapenémicos/uso terapéutico , Infección Hospitalaria , Farmacorresistencia Bacteriana , Sinergismo Farmacológico , Ertapenem , Infecciones por Escherichia coli/microbiología , Femenino , Humanos , Klebsiella pneumoniae/enzimología , Meropenem , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Infección de la Herida Quirúrgica , Tienamicinas , beta-Lactamasas/farmacología , beta-Lactamas
19.
J Clin Microbiol ; 53(1): 201-5, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25378572

RESUMEN

The Clinical and Laboratory Standards Institute (CLSI) lowered the MIC breakpoints for meropenem and imipenem from 4 mg/liter to 1 mg/liter for Enterobacteriaceae in 2010. The breakpoint change improves the probability of pharmacodynamic target attainment and eliminates the need for microbiology labs to perform confirmatory testing for Klebsiella pneumoniae carbapenemase (KPC) production or other beta-lactamases that hydrolyze carbapenems. However, there are limited data evaluating clinical outcomes of the affected breakpoints, and it is unknown if patients infected with Enterobacteriaceae with reduced susceptibility are more likely to have poor outcomes when treated with a carbapenem. We conducted a single-center retrospective matched-cohort analysis in adult patients with Enterobacteriaceae infections treated with meropenem, imipenem, or doripenem. Patients with Enterobacteriaceae infection with a carbapenem MIC of 2 to 8 mg/liter were matched based on pathogen, source of infection, comorbidities, and disease severity (1:1 ratio) to those with a carbapenem MIC of ≤1 mg/liter. A total of 36 patients were included in the study. The group with carbapenem MICs of 2 to 8 mg/liter had a significantly higher 30-day mortality than the group with carbapenem MICs of ≤1 mg/liter (38.9% compared to 5.6%, P = 0.04). Total hospital length of stay (LOS) and intensive care unit (ICU) LOS were longer in the group with MICs of 2 to 8 mg/liter than in the group with MICs of ≤1 mg/liter (57.6 days compared to 34.4 days [P = 0.06] and 56.6 days compared to 21.7 days [P < 0.01], respectively). Patients infected with Enterobacteriaceae with a carbapenem MIC of 2, 4, or 8 mg/liter had higher mortality rates and longer ICU LOS than matched cohorts with carbapenem MICs of ≤1 mg/liter, which supports CLSI's recommendation to lower susceptibility breakpoints for carbapenems.


Asunto(s)
Proteínas Bacterianas/farmacología , Proteínas Bacterianas/uso terapéutico , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Infecciones por Enterobacteriaceae/microbiología , Enterobacteriaceae/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Resistencia betalactámica , beta-Lactamasas/farmacología , beta-Lactamasas/uso terapéutico , Anciano , Comorbilidad , Infecciones por Enterobacteriaceae/epidemiología , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mortalidad , Estudios Retrospectivos , Factores de Riesgo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA