Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 18.696
Filter
1.
Euro Surveill ; 29(28)2024 Jul.
Article in English | MEDLINE | ID: mdl-38994602

ABSTRACT

Carbapenem-resistant Acinetobacter baumannii (CRAb) is an important pathogen causing serious nosocomial infections. We describe an outbreak of CRAb in an intensive care unit in the Netherlands in 2021. During an outbreak of non-resistant A. baumannii, while infection control measures were in place, CRAb isolates carrying highly similar bla NDM-1 - and tet(x3)-encoding plasmids were isolated from three patients over a period of several months. The chromosomal and plasmid sequences of the CRAb and non-carbapenemase-carrying A. baumannii isolates cultured from patient materials were analysed using hybrid assemblies of short-read and long-read sequences. The CRAb isolates revealed that the CRAb outbreak consisted of two different strains, carrying similar plasmids. The plasmids contained multiple antibiotic resistance genes including the tetracycline resistance gene tet(x3), and the bla NDM-1 and bla OXA-97 carbapenemase genes. We determined minimal inhibitory concentrations (MICs) for 13 antibiotics, including the newly registered tetracycline antibiotics eravacycline and omadacycline. The CRAb isolates showed high MICs for tetracycline antibiotics including eravacycline and omadacycline, except for minocycline which had a low MIC. In this study we show the value of sequencing multidrug-resistant A. baumannii for outbreak tracking and guiding outbreak mitigation measures.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Anti-Bacterial Agents , Cross Infection , Drug Resistance, Multiple, Bacterial , Microbial Sensitivity Tests , Tetracyclines , beta-Lactamases , Acinetobacter baumannii/genetics , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/isolation & purification , Acinetobacter baumannii/enzymology , Humans , Acinetobacter Infections/microbiology , Acinetobacter Infections/epidemiology , Tetracyclines/pharmacology , Anti-Bacterial Agents/pharmacology , Cross Infection/microbiology , Cross Infection/epidemiology , beta-Lactamases/genetics , Netherlands/epidemiology , Drug Resistance, Multiple, Bacterial/genetics , Plasmids/genetics , Disease Outbreaks , Bacterial Proteins/genetics , Carbapenems/pharmacology , Intensive Care Units
2.
BMC Infect Dis ; 24(1): 680, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982386

ABSTRACT

BACKGROUND: Aminoglycosides have been a cornerstone of the treatment of nosocomial infections caused by Pseudomonas aeruginosa for over 80 years. However, escalating emergence of resistance poses a significant challenge. Therefore, this study aimed to investigate the prevailing patterns of aminoglycoside resistance among clinical isolates of P. aeruginosa in Iran; as well as the underlying resistance mechanisms observed in patients referred to Ardabil hospitals. METHODS: A total of 200 isolates from five hospitals were evaluated. The resistance profiles of P. aeruginosa isolates to tobramycin, amikacin, and netilmicin were determined using the disk diffusion method. The capacity of aminoglycoside-resistant isolates to form biofilms was assessed through a phenotypic assay, and the results were confirmed using the gene amplification technique. The presence of genes associated with aminoglycoside resistance was detected using polymerase chain reaction (PCR). Quantitative reverse transcription PCR (qRT-PCR) was performed to measure the expression levels of genes encoding the MexXY-OprM efflux pump and PhoPQ two-component system (TCS). RESULTS: The prevalence of aminoglycoside-resistant P. aeruginosa isolates was 48%, with 94.7% demonstrating multidrug resistance (MDR). All aminoglycoside-resistant P. aeruginosa strains exhibited biofilm-forming capabilities and harbored all the genes associated with biofilm production. Among the nine genes encoding 16S rRNA methylase and aminoglycoside-modifying enzymes, three genes were detected in these isolates: aac(6')-Ib (85.4%), ant(2'')-Ia (18.7%), and aph(3')-VI (3.1%). Additionally, all aminoglycoside-resistant P. aeruginosa isolates carried mexY and phoP genes, although the expression levels of mexY and phoP were 75% and 87.5%, respectively. CONCLUSION: Given the considerably high prevalence of aminoglycoside-resistant P. aeruginosa strains, urgent measures are warranted to transition towards the use of novel aminoglycosides and to uphold vigilant surveillance of resistance patterns.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Biofilms , Microbial Sensitivity Tests , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/isolation & purification , Iran/epidemiology , Humans , Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Pseudomonas Infections/microbiology , Pseudomonas Infections/epidemiology , Biofilms/drug effects , Biofilms/growth & development , Prevalence , Drug Resistance, Multiple, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Resistance, Bacterial/genetics , Amikacin/pharmacology , Cross Infection/microbiology , Cross Infection/epidemiology , Tobramycin/pharmacology
3.
J Infect Dev Ctries ; 18(6): 937-942, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38990999

ABSTRACT

INTRODUCTION: Invasive device-associated nosocomial infections commonly occur in intensive care units (ICUs). These infections include intravascular catheter-related bloodstream infection (CRBSI), ventilator-associated pneumonia (VAP), and catheter-associated urinary tract infection (CAUTI). This study aimed to evaluate the factors associated with invasive device-associated nosocomial infections based on the underlying diseases of the patients and antibiotic resistance profiles of the pathogens causing the infections detected in the ICU in our hospital over a five-year period. METHODOLOGY: Invasive device-associated infections (CRBSI, VAP, and CAUTI) were detected retrospectively by the laboratory- and clinic-based active surveillance system according to the criteria of the US Centers for Disease Control and Prevention (CDC) in patients hospitalized in the ICU of the tertiary hospital between 1 January 2018 and 30 June 2023. RESULTS: A total of 425 invasive device-associated nosocomial infections and 441 culture results were detected (179 CRBSI, 176 VAP, 70 CAUTI). Out of them, 57 (13.4%) patients had hematological malignancy, 145 (34.1%) had solid organ malignancy, and 223 (52.5%) had no histopathologic diagnosis of any malignancy. An increase in extended-spectrum beta lactamase (ESBL) and carbapenem resistance in pathogens was detected during the study period. CONCLUSIONS: Antibiotic resistance of the Gram-negative bacteria associated with invasive device-associated infections increased during the study period. Antimicrobial stewardship will reduce rates of nosocomial infections, reduce mortality, and shorten hospital stay. Long-term catheterization and unnecessary antibiotic use should be avoided.


Subject(s)
Catheter-Related Infections , Cross Infection , Intensive Care Units , Pneumonia, Ventilator-Associated , Humans , Male , Retrospective Studies , Female , Cross Infection/microbiology , Cross Infection/epidemiology , Middle Aged , Catheter-Related Infections/microbiology , Catheter-Related Infections/epidemiology , Aged , Pneumonia, Ventilator-Associated/microbiology , Pneumonia, Ventilator-Associated/epidemiology , Adult , Urinary Tract Infections/microbiology , Urinary Tract Infections/epidemiology , Anti-Bacterial Agents/therapeutic use , Tertiary Care Centers/statistics & numerical data , Aged, 80 and over
4.
BMC Infect Dis ; 24(1): 662, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956542

ABSTRACT

BACKGROUND: Infections due to Citrobacter species are increasingly observed in hospitalized patients and are often multidrug-resistant. Yet, the magnitude and burden of Citrobacter spp. resistance in the hospital setting have not been reported. We aimed to evaluate the epidemiology of Citrobacter spp. infections among hospitalized patients, their main resistance patterns and Citrobacter spp. involvement in hospital outbreaks. METHODS: We conducted a systematic review and meta-analysis of published literature (PROSPERO registration Jan-2023, CRD42023390084). We searched Embase, Medline and grey literature for studies on hospitalized patients diagnosed with Citrobacter spp. infections, and nosocomial outbreaks due to Citrobacter spp. published during the years 2000-2022. We included observational, interventional, surveillance studies and outbreak reports. Outcomes of interest were the frequency of Citrobacter spp. infections among hospitalized patients and 3rd generation cephalosporin and/or carbapenem resistance percentages in these infections. We used random-effects models to generate pooled outcome estimates and evaluated risk of bias and quality of reporting of outbreaks. RESULTS: We screened 1609 deduplicated publications, assessed 148 full-texts, and included 41 studies (15 observational, 13 surveillance and 13 outbreak studies). Citrobacter spp. urinary tract- and bloodstream infections were most frequently reported, with Citrobacter freundii being the main causative species. Hospital-acquired infection occurred in 85% (838/990) of hospitalized patients with Citrobacter infection. After 2010, an increasing number of patients with Citrobacter spp. infections was reported in observational studies. Pooled frequency estimates for Citrobacter spp. infections could not be generated due to lack of data. The pooled prevalence of ESBL and carbapenemase producers among Citrobacter isolates were 22% (95%CI 4-50%, 7 studies) and 18% (95%CI 0-63%, 4 studies), respectively. An increased frequency of reported Citrobacter outbreaks was observed after 2016, with an infection/colonization ratio of 1:3 and a case-fatality ratio of 7% (6/89 patients). Common outbreak sources were sinks, toilets, contaminated food and injection material. Implemented preventive measures included environmental cleaning, isolation of positive patients and reinforcement of hand hygiene. Only seven out of 13 outbreaks (54%) were definitively controlled. CONCLUSION: This review highlights the clinical importance of endemic and epidemic Citrobacter spp. in healthcare settings. As an emerging, multidrug­resistant nosocomial pathogen it requires heightened awareness and further dedicated surveillance efforts.


Subject(s)
Citrobacter , Cross Infection , Enterobacteriaceae Infections , Humans , Enterobacteriaceae Infections/epidemiology , Enterobacteriaceae Infections/microbiology , Citrobacter/isolation & purification , Cross Infection/epidemiology , Cross Infection/microbiology , Hospitalization/statistics & numerical data , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Disease Outbreaks , Drug Resistance, Multiple, Bacterial , Urinary Tract Infections/epidemiology , Urinary Tract Infections/microbiology
5.
Tunis Med ; 102(7): 394-398, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38982962

ABSTRACT

INTRODUCTION: Healthcare-associated infections pose a significant public health burden, leading to morbidity, mortality, prolonged hospital stays, and substantial social and economic costs. Immunocompromised patients are at a heightened risk of nosocomial infections. AIM: This prospective study conducted at Mohammed VI University Hospital of Oujda aimed to assess the microbial ecology of surfaces and air in an immunosuppressed patient room compared to a double hospitalization room. METHODS: Microbiological air purity tests were conducted employing both the sedimentation method and the collision method with the assistance of Microflow Alpha. The sedimentation method used Mueller Hinton with 5% human blood, facilitating the free fall of contaminated dust particles. The collection program employed was set for 10 minutes per 1 m3. For surface sampling, swabs were taken from a 25 cm2 surface. The swabs were immediately forwarded to the Microbiology Laboratory. We carried out both macroscopic and microscopic identification of colonies, followed by definitive biochemical identification using the BD phoenixTM system. Antibiotic susceptibility was assessed through agar diffusion on Muller Hinton medium coupled with the determination of the minimum inhibitory concentration. RESULTS: The results revealed a decreased bacterial count within the protective isolation room, in contrast to the standard hospital room. We noted the predominance of coagulase-negative Staphylococcus spp and Bacillus spp. Staphylococcus aureus and Aspergillus spp, common pathogens in healthcare-associated infections, were notably absent in the protective isolation room. The findings underline the pivotal role of hospital environments in the transmission of healthcare-associated infections. CONCLUSION: The protective isolation room demonstrated effective control of microbial contamination, with fewer and less resistant germs. The study highlighted the significance of air treatment systems in preventing the spread of opportunistic infections. Our study underscored the critical role of microbiological cleanliness in preventing nosocomial infections.


Subject(s)
Air Microbiology , Cross Infection , Humans , Cross Infection/prevention & control , Cross Infection/epidemiology , Cross Infection/microbiology , Prospective Studies , Patients' Rooms/organization & administration , Patients' Rooms/statistics & numerical data , Patient Isolation/methods , Hospitals, University , Immunocompromised Host , Tunisia/epidemiology
6.
PLoS One ; 19(7): e0305083, 2024.
Article in English | MEDLINE | ID: mdl-38985740

ABSTRACT

Healthcare associated infections (HAIs) are costly but preventable. A limited understanding of the effects of environmental cleaning on the riskiest HAI associated pathogens is a current challenge in HAI prevention. This project aimed to quantify the effects of terminal hospital cleaning practices on HAI pathogens via environmental sampling in three hospitals located throughout the United States. Surfaces were swabbed from 36 occupied patient rooms with a laboratory-confirmed, hospital- or community-acquired infection of at least one of the four pathogens of interest (i.e., Acinetobacter baumannii (A. baumannii), methicillin resistant Staphylococcus aureus (MRSA), vancomycin resistant Enterococcus faecalis/faecium (VRE), and Clostridioides difficile (C. difficile)). Six nonporous, high touch surfaces (i.e., chair handrail, bed handrail, nurse call button, desk surface, bathroom counter near the sink, and a grab bar near the toilet) were sampled in each room for Adenosine Triphosphate (ATP) and the four pathogens of interest before and after terminal cleaning. The four pathogens of interest were detected on surfaces before and after terminal cleaning, but their levels were generally reduced. Overall, C. difficile was confirmed on the desk (n = 2), while MRSA (n = 24) and VRE (n = 25) were confirmed on all surface types before terminal cleaning. After cleaning, only MRSA (n = 6) on bed handrail, chair handrail, and nurse call button and VRE (n = 5) on bathroom sink, bed handrail, nurse call button, toilet grab bar, and C. difficile (n = 1) were confirmed. At 2 of the 3 hospitals, pathogens were generally reduced by >99% during terminal cleaning. One hospital showed that VRE increased after terminal cleaning, MRSA was reduced by 73% on the nurse call button, and VRE was reduced by only 50% on the bathroom sink. ATP detections did not correlate with any pathogen concentration. This study highlights the importance of terminal cleaning and indicates room for improvement in cleaning practices to reduce surface contamination throughout hospital rooms.


Subject(s)
Clostridioides difficile , Cross Infection , Methicillin-Resistant Staphylococcus aureus , Patients' Rooms , Cross Infection/microbiology , Cross Infection/prevention & control , Humans , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Clostridioides difficile/isolation & purification , Housekeeping, Hospital , Acinetobacter baumannii/isolation & purification , Infection Control/methods , Vancomycin-Resistant Enterococci/isolation & purification
7.
Front Cell Infect Microbiol ; 14: 1335096, 2024.
Article in English | MEDLINE | ID: mdl-38975326

ABSTRACT

Objective: Pseudomonas aeruginosa, a difficult-to-manage nosocomial pathogen, poses a serious threat to clinical outcomes in intensive care (ICU) patients due to its high antimicrobial resistance (AMR). To promote effective management, it is essential to investigate the genomic and phenotypic differences in AMR expression of the isolates. Methods: A prospective observational study was conducted from July 2022 to April 2023 at Liepaja Regional Hospital in Latvia. The study included all adult patients who were admitted to the ICU and had a documented infection with P. aeruginosa, as confirmed by standard laboratory microbiological testing and short-read sequencing. Since ResFinder is the only sequencing-based database offering antibacterial susceptibility testing (AST) data for each antibiotic, we conducted a comparison of the resistance profile with the results of phenotypic testing, evaluating if ResFinder met the US Food and Drug Administration (FDA) requirements for approval as a new AMR diagnostic test. Next, to improve precision, AST data from ResFinder was compared with two other databases - AMRFinderPlus and RGI. Additionally, data was gathered from environmental samples to inform the implementation of appropriate infection control measures in real time. Results: Our cohort consisted of 33 samples from 29 ICU patients and 34 environmental samples. The presence of P. aeruginosa infection was found to be associated with unfavourable clinical outcomes. A third of the patient samples were identified as multi-drug resistant isolates. Apart from resistance against colistin, significant discrepancies were observed when phenotypic data were compared to genotypic data. For example, the aminoglycoside resistance prediction of ResFinder yielded a major errors value of 3.03% for amikacin, which was marginally above the FDA threshold. Among the three positive environmental samples, one sample exhibited multiple AMR genes similar to the patient samples in its cluster. Conclusion: Our findings underscore the importance of utilizing a combination of diagnostic methods for the identification of resistance mechanisms, clusters, and environmental reservoirs in ICUs.


Subject(s)
Anti-Bacterial Agents , Intensive Care Units , Microbial Sensitivity Tests , Phenotype , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Humans , Pseudomonas Infections/microbiology , Anti-Bacterial Agents/pharmacology , Prospective Studies , Female , Male , Middle Aged , Cross Infection/microbiology , Aged , Drug Resistance, Bacterial/genetics , Drug Resistance, Multiple, Bacterial/genetics , Genomics/methods , Latvia , Adult , Colistin/pharmacology , Genome, Bacterial/genetics
8.
Microb Genom ; 10(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38967541

ABSTRACT

Outbreaks of methicillin-resistant Staphylococcus aureus (MRSA) are well described in the neonatal intensive care unit (NICU) setting. Genomics has revolutionized the investigation of such outbreaks; however, to date, this has largely been completed retrospectively and has typically relied on short-read platforms. In 2022, our laboratory established a prospective genomic surveillance system using Oxford Nanopore Technologies sequencing for rapid outbreak detection. Herein, using this system, we describe the detection and control of an outbreak of sequence-type (ST)97 MRSA in our NICU. The outbreak was identified 13 days after the first MRSA-positive culture and at a point where there were only two known cases. Ward screening rapidly defined the extent of the outbreak, with six other infants found to be colonized. There was minimal transmission once the outbreak had been detected and appropriate infection control measures had been instituted; only two further ST97 cases were detected, along with three unrelated non-ST97 MRSA cases. To contextualize the outbreak, core-genome single-nucleotide variants were identified for phylogenetic analysis after de novo assembly of nanopore data. Comparisons with global (n=45) and national surveillance (n=35) ST97 genomes revealed the stepwise evolution of methicillin resistance within this ST97 subset. A distinct cluster comprising nine of the ten ST97-IVa genomes from the NICU was identified, with strains from 2020 to 2022 national surveillance serving as outgroups to this cluster. One ST97-IVa genome presumed to be part of the outbreak formed an outgroup and was retrospectively excluded. A second phylogeny was created using Illumina sequencing, which considerably reduced the branch lengths of the NICU isolates on the phylogenetic tree. However, the overall tree topology and conclusions were unchanged, with the exception of the NICU outbreak cluster, where differences in branch lengths were observed. This analysis demonstrated the ability of a nanopore-only prospective genomic surveillance system to rapidly identify and contextualize an outbreak of MRSA in a NICU.


Subject(s)
Disease Outbreaks , Intensive Care Units, Neonatal , Methicillin-Resistant Staphylococcus aureus , Nanopore Sequencing , Phylogeny , Staphylococcal Infections , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/classification , Humans , Staphylococcal Infections/epidemiology , Staphylococcal Infections/microbiology , Infant, Newborn , Nanopore Sequencing/methods , Cross Infection/epidemiology , Cross Infection/microbiology , Prospective Studies , Genome, Bacterial , Polymorphism, Single Nucleotide , Female
9.
Antimicrob Resist Infect Control ; 13(1): 70, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961463

ABSTRACT

OBJECTIVES: Genomic surveillance of Klebsiella pneumoniae carbapenemase-producing Klebsiella pneumoniae (KPC-Kp) is crucial for virulence, drug-resistance monitoring, and outbreak containment. METHODS: Genomic analysis on 87 KPC-Kp strains isolated from 3 Northern Italy hospitals in 2019-2021 was performed by whole genome sequencing (WGS), to characterize resistome, virulome, and mobilome, and to assess potential associations with phenotype resistance and clinical presentation. Maximum Likelihood and Minimum Spanning Trees were used to determine strain correlations and identify potential transmission clusters. RESULTS: Overall, 15 different STs were found; the predominant ones included ST307 (35, 40.2%), ST512/1519 (15, 17.2%), ST20 (12, 13.8%), and ST101 (7, 8.1%). 33 (37.9%) KPC-Kp strains were noticed to be in five transmission clusters (median number of isolates in each cluster: 5 [3-10]), four of them characterized by intra-hospital transmission. All 87 strains harbored Tn4401a transposon, carrying blaKPC-3 (48, 55.2%), blaKPC-2 (38, 43.7%), and in one case (1.2%) blaKPC-33, the latter gene conferred resistance to ceftazidime/avibactam (CZA). Thirty strains (34.5%) harbored porin mutations; of them, 7 (8.1%) carried multiple Tn4401a copies. These strains were characterized by significantly higher CZA minimum inhibitory concentration compared with strains with no porin mutations or single Tn4401a copy, respectively, even if they did not overcome the resistance breakpoint of 8 ug/mL. Median 2 (IQR:1-2) virulence factors per strain were detected. The lowest number was observed in ST20 compared to the other STs (p<0.001). While ST307 was associated with infection events, a trend associated with colonization events could be observed for ST20. CONCLUSIONS: Integration of genomic, resistance score, and clinical data allowed us to define a relative diversification of KPC-Kp in Northern Italy between 2019 and 2021, characterized by few large transmission chains and rare inter-hospital transmission. Our results also provided initial evidence of correlation between KPC-Kp genomic signatures and higher MIC levels to some antimicrobial agents or colonization/infection status, once again underlining WGS's importance in bacterial surveillance.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Hospitals, University , Klebsiella Infections , Klebsiella pneumoniae , Microbial Sensitivity Tests , Whole Genome Sequencing , beta-Lactamases , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/isolation & purification , Klebsiella pneumoniae/classification , Klebsiella pneumoniae/enzymology , Italy/epidemiology , Humans , beta-Lactamases/genetics , Klebsiella Infections/microbiology , Klebsiella Infections/epidemiology , Bacterial Proteins/genetics , Anti-Bacterial Agents/pharmacology , Genome, Bacterial , Drug Resistance, Multiple, Bacterial/genetics , Genomics , Cross Infection/microbiology , Cross Infection/epidemiology
10.
Mycoses ; 67(7): e13765, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38988310

ABSTRACT

BACKGROUND: Candida auris, a multidrug-resistant fungal pathogen, has received considerable attention owing to its recent surge, especially in South America, which coincides with the ongoing global COVID-19 pandemic. Understanding the clinical and microbiological characteristics of outbreaks is crucial for their effective management and control. OBJECTIVE: This retrospective observational study aimed to characterize a C. auris outbreak at a Peruvian referral hospital between January 2021 and July 2023. METHODS: Data were collected from hospitalized patients with positive C. auris culture results. Microbiological data and antifungal susceptibility test results were analysed. Additionally, infection prevention and control measures have been described. Statistical analysis was used to compare the characteristics between the infected and colonized patients. RESULTS: Thirty-three patients were identified, mostly male (66.7%), with a median age of 53 years. Among them, 18 (54.5%) were colonized, and 15 (45.5%) were infected. Fungemia was the predominant presentation (80%), with notable cases of fungemia in tuberculosis patients with long-stay devices for parenteral anti-tuberculosis therapy. Seventy-five percent of the isolates exhibited fluconazole resistance. Echinocandins were the primary treatment, preventing fungemia recurrence within 30 days. Infected patients had significantly longer hospital stays than colonized patients (100 vs. 45 days; p = .023). Hospital mortality rates were 46.7% and 25% in the infected and fungemia patients, respectively. Simultaneous outbreaks of multidrug-resistant bacteria were documented. CONCLUSIONS: This study underscores the severity of a C. auris outbreak at a referral hospital in Peru, highlighting its significant impact on patient outcomes and healthcare resources. The high prevalence of fluconazole-resistant isolates, leading to prolonged hospital stay and high mortality rates, particularly in cases of fungemia, underscores the critical need for effective infection prevention and control strategies.


Subject(s)
Antifungal Agents , Candida auris , Candidiasis , Disease Outbreaks , Humans , Peru/epidemiology , Middle Aged , Male , Female , Retrospective Studies , Adult , Candidiasis/epidemiology , Candidiasis/microbiology , Antifungal Agents/therapeutic use , Antifungal Agents/pharmacology , Aged , Candida auris/drug effects , COVID-19/epidemiology , Microbial Sensitivity Tests , Cross Infection/epidemiology , Cross Infection/microbiology , Candida/drug effects , Candida/isolation & purification , Candida/classification , Referral and Consultation
11.
Microbiol Spectr ; 12(7): e0394723, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38864670

ABSTRACT

Clostridioides difficile (C. difficile) is widely distributed in the intestinal tract of humans, animals, and in the environment. It is the most common cause of diarrhea associated with the use of antimicrobials in humans and among the most common healthcare-associated infections worldwide. Its pathogenesis is mainly due to the production of toxin A (TcdA), toxin B (TcdB), and a binary toxin (CDT), whose genetic variants may be associated with disease severity. We studied genetic diversity in 39 C. difficile isolates from adults and children attended at two Mexican hospitals, using different gene and genome typing methods and investigated their association with in vitro expression of toxins. Whole-genome sequencing in 39 toxigenic C. difficile isolates were used for multilocus sequence typing, tcdA, and tcdB typing sequence type, and phylogenetic analysis. Strains were grown in broth media, and expression of toxin genes was measured by real-time PCR and cytotoxicity in cell-culture assays. Clustering of strains by genome-wide phylogeny matched clade classification, forming different subclusters within each clade. The toxin profile tcdA+/tcdB+/cdt+ and clade 2/ST1 were the most prevalent among isolates from children and adults. Isolates presented two TcdA and three TcdB subtypes, of which TcdA2 and TcdB2 were more prevalent. Prevalent clades and toxin subtypes in strains from children differed from those in adult strains. Toxin gene expression or cytotoxicity was not associated with genotyping or toxin subtypes. In conclusion, genomic and phenotypic analysis shows high diversity among C. difficile isolates from patients with healthcare-associated diarrhea. IMPORTANCE: Clostridioides difficile is a toxin-producing bacterial pathogen recognized as the most common cause of diarrhea acquired primarily in healthcare settings. This bacterial species is diverse; its global population has been divided into five different clades using multilocus sequence typing, and strains may express different toxin subtypes that may be related to the clades and, importantly, to the severity and progression of disease. Genotyping of children strains differed from adults suggesting toxins might present a reduced toxicity. We studied extensively cytotoxicity, expression of toxins, whole genome phylogeny, and toxin typing in clinical C. difficile isolates. Most isolates presented a tcdA+/ tcdB+/cdt+ pattern, with high diversity in cytotoxicity and clade 2/ST1 was the most prevalent. However, they all had the same TcdA2/TcdB2 toxin subtype. Advances in genomics and bioinformatics tools offer the opportunity to understand the virulence of C. difficile better and find markers for better clinical use.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Cross Infection , Diarrhea , Genetic Variation , Multilocus Sequence Typing , Phylogeny , Humans , Clostridioides difficile/genetics , Clostridioides difficile/classification , Clostridioides difficile/isolation & purification , Diarrhea/microbiology , Diarrhea/epidemiology , Mexico/epidemiology , Child , Bacterial Toxins/genetics , Adult , Clostridium Infections/microbiology , Clostridium Infections/epidemiology , Cross Infection/microbiology , Cross Infection/epidemiology , Bacterial Proteins/genetics , Enterotoxins/genetics , Male , Child, Preschool , Female , Prevalence , Adolescent , Whole Genome Sequencing , Phenotype , Genome, Bacterial/genetics , Infant , Middle Aged , Genomics
12.
J Infect Public Health ; 17(7): 102463, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38833914

ABSTRACT

BACKGROUND: Enterococcus avium (E. avium) is a Gram-positive nosocomial pathogen that is commonly isolated from the alimentary tract. The objective of this functional genomics study was to identify the resistant genes by analyzing the genome of E. avium IRMC1622a, a type of bacteria found in feces collected from a patient at a Saudi Arabian tertiary hospital. METHODS: The bacterial strain IRMC1622a was identified by 16 S rRNA sequencing as Enterococcus sp. The resistance phenomics were performed using VITEK® 2, and morphological analysis was achieved using a scanning electron microscope (SEM). Finally, the whole bacterial genome of the bacterial strain IRMC1622a was subjected to sequencing during October 2023 using Oxford Nanopore long-read sequencing technology, and mining for resistant genes. RESULTS: The results of antimicrobial resistant phenomics indicated that the IRMC1622a strain was sensitive to all tested antimicrobial agents except for erythromycin, and the same result was confirmed by genomic analysis in addition to other classes of antibiotics. SEM showed E. avium IRMC1622a is ovoid shape, in single cells (L 1.2797 ± 0.1490 µm), in pairs (L 1.7333 ± 0.1054 µm), and in chains (L 2.44033 ± 0.1978 µm). The E. avium IRMC1622a genome has 14 (in CARD) antimicrobial resistance genes that were identified with several mechanisms of antimicrobial resistance, such as the efflux pump and conferring antibiotic resistance. The present study revealed that the E. avium IRMC1622a genome contains a high number of genes associated with virulence factors, and 14 matched pathogenic protein families and predicted as human pathogen (probability score 0.855). We report two (ISEnfa4 and ISEfa5) mobile genetic elements for the first time in the E. avium genome. CONCLUSIONS: The study concludes that E. avium IRMC1622a is susceptible to all tested antibacterials except erythromycin. The IRMC1622a has 14 genes encoding antimicrobial resistance mechanisms, including the efflux pump and conferring antibiotic resistance. This could indicate a potential rise in E. avium resistance in healthcare facilities. These observations may raise concerns regarding E. avium resistance in healthcare. We need more research to understand the pathophysiology of E. avium, which leads to hospital-acquired infections.


Subject(s)
Anti-Bacterial Agents , Feces , Genome, Bacterial , Microbial Sensitivity Tests , Humans , Anti-Bacterial Agents/pharmacology , Feces/microbiology , Gram-Positive Bacterial Infections/microbiology , Genomics , Saudi Arabia , Enterococcus/genetics , Enterococcus/drug effects , Enterococcus/isolation & purification , RNA, Ribosomal, 16S/genetics , Drug Resistance, Bacterial/genetics , Whole Genome Sequencing , Tertiary Care Centers , Cross Infection/microbiology , Phenotype
13.
J Infect Public Health ; 17(7): 102469, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38838607

ABSTRACT

BACKGROUND: In recent years, the global spread of antimicrobial resistance has become a concerning issue, often referred to as a "silent pandemic". Healthcare-associated infections (HAIs) caused by antibiotic-resistant bacteria (ARB) are a recurring problem, with some originating from waterborne route. The study aimed to investigate the presence of clinically relevant opportunistic bacteria and antibiotic resistance genes (ARGs) in hospital water distribution systems (WDSs). METHODS: Water and biofilm samples (n = 192) were collected from nine hospitals in Isfahan and Kashan, located in central Iran, between May 2022 and June 2023. The samples were analyzed to determine the presence and quantities of opportunistic bacteria and ARGs using cultural and molecular methods. RESULTS: Staphylococcus spp. were highly detected in WDS samples (90 isolates), with 33 % of them harboring mecA gene. However, the occurrences of E. coli (1 isolate), Acinetobacter baumannii (3 isolates), and Pseudomonas aeruginosa (14 isolates) were low. Moreover, several Gram-negative bacteria containing ARGs were identified in the samples, mainly belonging to Stenotrophomonas, Sphingomonas and Brevundimonas genera. Various ARGs, as well as intI1, were found in hospital WDSs (ranging from 14 % to 60 %), with higher occurrences in the biofilm samples. CONCLUSION: Our results underscore the importance of biofilms in water taps as hotspots for the dissemination of opportunistic bacteria and ARG within hospital environments. The identification of multiple opportunistic bacteria and ARGs raises concerns about the potential exposure and acquisition of HAIs, emphasizing the need for proactive measures, particularly in controlling biofilms, to mitigate infection risks in healthcare settings.


Subject(s)
Biofilms , Cross Infection , Hospitals , Biofilms/drug effects , Biofilms/growth & development , Cross Infection/microbiology , Humans , Iran/epidemiology , Drug Resistance, Bacterial/genetics , Water Microbiology , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/classification , Anti-Bacterial Agents/pharmacology
14.
BMC Microbiol ; 24(1): 215, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38890594

ABSTRACT

BACKGROUND: A multidrug-resistant lineage of Staphylococcus epidermidis named ST215 is a common cause of prosthetic joint infections and other deep surgical site infections in Northern Europe, but is not present elsewhere. The increasing resistance among S. epidermidis strains is a global concern. We used whole-genome sequencing to characterize ST215 from healthcare settings. RESULTS: We completed the genome of a ST215 isolate from a Swedish hospital using short and long reads, resulting in a circular 2,676,787 bp chromosome and a 2,326 bp plasmid. The new ST215 genome was placed in phylogenetic context using 1,361 finished public S. epidermidis reference genomes. We generated 10 additional short-read ST215 genomes and 11 short-read genomes of ST2, which is another common multidrug-resistant lineage at the same hospital. We studied recombination's role in the evolution of ST2 and ST215, and found multiple recombination events averaging 30-50 kb. By comparing the results of antimicrobial susceptibility testing for 31 antimicrobial drugs with the genome content encoding antimicrobial resistance in the ST215 and ST2 isolates, we found highly similar resistance traits between the isolates, with 22 resistance genes being shared between all the ST215 and ST2 genomes. The ST215 genome contained 29 genes that were historically identified as virulence genes of S. epidermidis ST2. We established that in the nucleotide sequence stretches identified as recombination events, virulence genes were overrepresented in ST215, while antibiotic resistance genes were overrepresented in ST2. CONCLUSIONS: This study features the extensive antibiotic resistance and virulence gene content in ST215 genomes. ST215 and ST2 lineages have similarly evolved, acquiring resistance and virulence through genomic recombination. The results highlight the threat of new multidrug-resistant S. epidermidis lineages emerging in healthcare settings.


Subject(s)
Anti-Bacterial Agents , Cross Infection , Drug Resistance, Multiple, Bacterial , Genome, Bacterial , Phylogeny , Staphylococcal Infections , Staphylococcus epidermidis , Whole Genome Sequencing , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/isolation & purification , Staphylococcus epidermidis/pathogenicity , Drug Resistance, Multiple, Bacterial/genetics , Genome, Bacterial/genetics , Humans , Staphylococcal Infections/microbiology , Cross Infection/microbiology , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Sweden , Plasmids/genetics , Recombination, Genetic
15.
Antimicrob Resist Infect Control ; 13(1): 65, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38886759

ABSTRACT

BACKGROUND: Stenotrophomonas maltophilia, a multidrug-resistant gram-negative bacteria (GNB), is an emerging nosocomial pathogen. This study assessed the clinical outcomes of GNB infections in surgical intensive care unit (SICU) patients post-abdominal surgery, focusing on the differences between S. maltophilia and other GNBs, including Pseudomonas aeruginosa. METHODS: A retrospective study was conducted on SICU patients at Kaohsiung Chang Gung Memorial Hospital from 2010 to 2020, who developed GNB infections following abdominal surgery. RESULTS: Of 442 patients, 237 had S. maltophilia and 205 had non-S. maltophilia GNB infections (including 81 with P. aeruginosa). The overall mortality rate was 44.5%, and S. maltophilia infection emerged as a significant contributor to the mortality rate in patients with GNB infections. S. maltophilia patients had longer mechanical ventilation and SICU stays, with a 30-day mortality rate of 35.4%, higher than the non-S. maltophilia GNB (22.9%) and P. aeruginosa (21%) groups. In-hospital mortality was also higher in the S. maltophilia group (53.2%) compared to the non-S. maltophilia GNB (34.6%) and P. aeruginosa groups (29.6%). Risk factors for acquiring S. maltophilia included a higher Sequential Organ Failure Assessment score and prior broad-spectrum antibiotics use. Older age, polymicrobial infections, and elevated bilirubin were associated with increased 30-day mortality in S. maltophilia patients. CONCLUSION: S. maltophilia infections in post-abdominal surgery patients are linked to higher mortality than non-S. maltophilia GNB and P. aeruginosa infections, emphasizing the need for early diagnosis and treatment to improve outcomes.


Subject(s)
Gram-Negative Bacterial Infections , Intensive Care Units , Stenotrophomonas maltophilia , Humans , Gram-Negative Bacterial Infections/mortality , Male , Retrospective Studies , Female , Middle Aged , Aged , Abdomen/surgery , Hospital Mortality , Pseudomonas aeruginosa , Adult , Cross Infection/mortality , Cross Infection/microbiology , Anti-Bacterial Agents/therapeutic use
16.
Euro Surveill ; 29(26)2024 Jun.
Article in English | MEDLINE | ID: mdl-38940004

ABSTRACT

In 2022, an outbreak with severe bloodstream infections caused by Serratia marcescens occurred in an adult intensive care unit (ICU) in Hungary. Eight cases, five of whom died, were detected. Initial control measures could not stop the outbreak. We conducted a matched case-control study. In univariable analysis, the cases were more likely to be located around one sink in the ICU and had more medical procedures and medications than the controls, however, the multivariable analysis was not conclusive. Isolates from blood cultures of the cases and the ICU environment were closely related by whole genome sequencing and resistant or tolerant against the quaternary ammonium compound surface disinfectant used in the ICU. Thus, S. marcescens was able to survive in the environment despite regular cleaning and disinfection. The hospital replaced the disinfectant with another one, tightened the cleaning protocol and strengthened hand hygiene compliance among the healthcare workers. Together, these control measures have proved effective to prevent new cases. Our results highlight the importance of multidisciplinary outbreak investigations, including environmental sampling, molecular typing and testing for disinfectant resistance.


Subject(s)
Cross Infection , Disease Outbreaks , Disinfectants , Intensive Care Units , Serratia Infections , Serratia marcescens , Humans , Serratia marcescens/drug effects , Serratia marcescens/genetics , Serratia marcescens/isolation & purification , Cross Infection/epidemiology , Cross Infection/microbiology , Hungary/epidemiology , Serratia Infections/epidemiology , Serratia Infections/microbiology , Disinfectants/pharmacology , Case-Control Studies , Male , Female , Adult , Middle Aged , Whole Genome Sequencing , Disinfection/methods , Aged , Infection Control/methods , Drug Resistance, Bacterial
17.
Microb Genom ; 10(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38896471

ABSTRACT

Carbapenems are last-resort antibiotics for treatment of infections caused by multidrug-resistant Enterobacterales, but carbapenem resistance is a rising global threat due to the acquisition of carbapenemase genes. Oxacillinase-48 (bla OXA-48)-type carbapenemases are increasing in abundance in Canada and elsewhere; these genes are frequently found on mobile genetic elements and are associated with specific transposons. This means that alongside clonal dissemination, bla OXA-48-type genes can spread through plasmid-mediated horizontal gene transfer. We applied whole genome sequencing to characterize 249 bla OXA-48-type-producing Enterobacterales isolates collected by the Canadian Nosocomial Infection Surveillance Program from 2010 to 2021. Using a combination of short- and long-read sequencing, we obtained 70 complete and circular bla OXA-48-type-encoding plasmids. Using MOB-suite, four major plasmids clustered were identified, and we further estimated a plasmid cluster for 91.9 % (147/160) of incomplete bla OXA-48-type-encoding contigs. We identified different patterns of carbapenemase mobilization across Canada, including horizontal transmission of bla OXA-181/IncX3 plasmids (75/249, 30.1 %) and bla OXA-48/IncL/M plasmids (47/249, 18.9 %), and both horizontal transmission and clonal transmission of bla OXA-232 for Klebsiella pneumoniae ST231 on ColE2-type/ColKP3 plasmids (25/249, 10.0 %). Our findings highlight the diversity of OXA-48-type plasmids and indicate that multiple plasmid clusters and clonal transmission have contributed to bla OXA-48-type spread and persistence in Canada.


Subject(s)
Bacterial Proteins , Carbapenems , Enterobacteriaceae Infections , Plasmids , Whole Genome Sequencing , beta-Lactamases , beta-Lactamases/genetics , Plasmids/genetics , Canada/epidemiology , Humans , Carbapenems/pharmacology , Bacterial Proteins/genetics , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/epidemiology , Enterobacteriaceae/genetics , Enterobacteriaceae/drug effects , Enterobacteriaceae/classification , Gene Transfer, Horizontal , Anti-Bacterial Agents/pharmacology , Cross Infection/microbiology , Cross Infection/epidemiology
18.
BMC Microbiol ; 24(1): 225, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926687

ABSTRACT

BACKGROUND: The incidence of hospital-acquired infections in extensively drug-resistant Pseudomonas aeruginosa (XDR-PA) has been increasing worldwide and is frequently associated with an increase in mortality and morbidity rates. The aim of this study was to characterize clinical XDR-PA isolates recovered during six months at three different hospitals in Egypt. RESULTS: Seventy hospital-acquired clinical isolates of P. aeruginosa were classified into multidrug-resistant (MDR), extensively drug-resistant (XDR) and pandrug-resistant (PDR), according to their antimicrobial resistance profile. In addition, the possession of genes associated with mobile genetic elements and genes encoding antimicrobial resistance determinants among isolates were detected using polymerase chain reaction. As a result, a significant percentage of the isolates (75.7%) were XDR, while 18.5% were MDR, however only 5.7% of the isolates were non-MDR. The phenotypic detection of carbapenemases, extended-spectrum ß-lactamases (ESBLs) and metallo ß-lactamase (MBL) enzymes showed that 73.6% of XDR-PA isolates were carbapenemases producers, whereas 75.5% and 88.7% of XDR-PA isolates produced ESBLs and MBL respectively. In addition, PCR screening showed that oxa gene was the most frequently detected gene of carbapenemases (91.4%), while aac(6')-lb gene was mostly detected (84.3%) among the screened aminoglycosides-resistance genes. Furthermore, the molecular detection of the colistin resistance gene showed that 12.9% of isolates harbored mcr-1 gene. Concerning mobile genetic element markers (intI, traA, tnp513, and merA), intI was the highest detected gene as it was amplified in 67 isolates (95.7%). Finally, phylogenetic and molecular typing of the isolates via ERIC-PCR analysis revealed 10 different ERIC fingerprints. CONCLUSION: The present study revealed a high prevalence of XDR-PA in hospital settings which were resistant to a variety of antibiotics due to several mechanisms. In addition, 98% of the XDR-PA clinical isolates contained at least one gene associated with movable genetic elements, which could have aided the evolution of these XDR-PA strains. To reduce spread of drug resistance, judicious use of antimicrobial agents and strict infection control measures are therefore essential.


Subject(s)
Anti-Bacterial Agents , Cross Infection , Drug Resistance, Multiple, Bacterial , Microbial Sensitivity Tests , Pseudomonas Infections , Pseudomonas aeruginosa , beta-Lactamases , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/isolation & purification , Humans , Pseudomonas Infections/microbiology , Pseudomonas Infections/epidemiology , Drug Resistance, Multiple, Bacterial/genetics , Cross Infection/microbiology , Cross Infection/epidemiology , Egypt/epidemiology , beta-Lactamases/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Hospitals/statistics & numerical data , Interspersed Repetitive Sequences/genetics , Polymerase Chain Reaction
19.
PLoS One ; 19(6): e0304378, 2024.
Article in English | MEDLINE | ID: mdl-38865328

ABSTRACT

OBJECTIVE: Evaluate the effects of five disinfection methods on bacterial concentrations in hospital sink drains, focusing on three opportunistic pathogens (OPs): Serratia marcescens, Pseudomonas aeruginosa and Stenotrophomonas maltophilia. DESIGN: Over two years, three sampling campaigns were conducted in a neonatal intensive care unit (NICU). Samples from 19 sink drains were taken at three time points: before, during, and after disinfection. Bacterial concentration was measured using culture-based and flow cytometry methods. High-throughput short sequence typing was performed to identify the three OPs and assess S. marcescens persistence after disinfection at the genotypic level. SETTING: This study was conducted in a pediatric hospitals NICU in Montréal, Canada, which is divided in an intensive and intermediate care side, with individual rooms equipped with a sink. INTERVENTIONS: Five treatments were compared: self-disinfecting drains, chlorine disinfection, boiling water disinfection, hot tap water flushing, and steam disinfection. RESULTS: This study highlights significant differences in the effectiveness of disinfection methods. Chlorine treatment proved ineffective in reducing bacterial concentration, including the three OPs. In contrast, all other drain interventions resulted in an immediate reduction in culturable bacteria (4-8 log) and intact cells (2-3 log). Thermal methods, particularly boiling water and steam treatments, exhibited superior effectiveness in reducing bacterial loads, including OPs. However, in drains with well-established bacterial biofilms, clonal strains of S. marcescens recolonized the drains after heat treatments. CONCLUSIONS: Our study supports thermal disinfection (>80°C) for pathogen reduction in drains but highlights the need for additional trials and the implementation of specific measures to limit biofilm formation.


Subject(s)
Disinfection , Intensive Care Units, Neonatal , Serratia marcescens , Serratia marcescens/drug effects , Disinfection/methods , Humans , Pseudomonas aeruginosa/drug effects , Infant, Newborn , Stenotrophomonas maltophilia/drug effects , Serratia Infections/microbiology , Serratia Infections/prevention & control , Cross Infection/prevention & control , Cross Infection/microbiology
20.
ACS Infect Dis ; 10(7): 2336-2355, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38866389

ABSTRACT

The misuse of antibiotics has led to the global spread of drug-resistant bacteria, especially multi-drug-resistant (MDR) ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species). These opportunistic bacteria pose a significant threat, in particular within hospitals, where they cause nosocomial infections, leading to substantial morbidity and mortality. To comprehensively explore ESKAPE pathogenesis, virulence, host immune response, diagnostics, and therapeutics, researchers increasingly rely on necessitate suitable animal infection models. However, no single model can fully replicate all aspects of infectious diseases. Notably when studying opportunistic pathogens in immunocompetent hosts, rapid clearance by the host immune system can limit the expression of characteristic disease symptoms. In this study, we examine the critical role of animal infection models in understanding ESKAPE pathogens, addressing limitations and research gaps. We discuss applications and highlight key considerations for effective models. Thoughtful decisions on disease replication, parameter monitoring, and data collection are crucial for model reliability. By meticulously replicating human diseases and addressing limitations, researchers maximize the potential of animal infection models. This aids in targeted therapeutic development, bridges knowledge gaps, and helps combat MDR ESKAPE pathogens, safeguarding public health.


Subject(s)
Disease Models, Animal , Drug Resistance, Multiple, Bacterial , Animals , Humans , Enterococcus faecium/drug effects , Enterococcus faecium/physiology , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Klebsiella pneumoniae/drug effects , Acinetobacter baumannii/drug effects , Cross Infection/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Enterobacter/drug effects , Bacterial Infections/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...