Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Antimicrob Agents Chemother ; 59(8): 4533-43, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25987626

RESUMO

Asymptomatic colonization may contribute to Clostridium difficile transmission. Few data identify which patients are at risk for colonization. We performed a prospective cohort study of C. difficile colonization and risk factors for C. difficile acquisition and loss in hospitalized patients. Patients admitted to medical or surgical wards at a tertiary care hospital were enrolled; interviews and chart review were performed to determine patient demographics, C. difficile infection (CDI) history, medications, and health care exposures. Stool samples/rectal swabs were collected at enrollment and discharge; stool samples from clinical laboratory tests were also included. Samples were cultured for C. difficile, and the isolates were tested for toxins A and B and ribotyped. Chi-square tests and univariate logistic regression were used for the analyses. Two hundred thirty-five patients were enrolled. Of the patients, 21% were colonized with C. difficile (toxigenic and nontoxigenic) at admission and 24% at discharge. Ribotype 027 accounted for 6% of the strains at admission and 12% at discharge. Of the patients colonized at admission, 78% were also colonized at discharge. Cephalosporin use was associated with C. difficile acquisition (47% of patients who acquired C. difficile versus 25% of patients who did not; P = 0.03). ß-lactam-ß-lactamase inhibitor combinations were associated with a loss of C. difficile colonization (36% of patients who lost C. difficile colonization versus 8% of patients colonized at both admission and discharge; P = 0.04), as was metronidazole (27% versus 3%; P = 0.03). Antibiotic use affects the epidemiology of asymptomatic C. difficile colonization, including acquisition and loss, and it requires additional study.


Assuntos
Clostridioides difficile/isolamento & purificação , Infecções por Clostridium/microbiologia , Antibacterianos/uso terapêutico , Cefalosporinas/uso terapêutico , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/tratamento farmacológico , Infecção Hospitalar/tratamento farmacológico , Infecção Hospitalar/microbiologia , Fezes/microbiologia , Feminino , Hospitalização , Hospitais , Humanos , Modelos Logísticos , Masculino , Estudos Prospectivos , Ribotipagem/métodos , Fatores de Risco , Inibidores de beta-Lactamases/uso terapêutico
2.
Clin Infect Dis ; 59(2): 216-22, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24755858

RESUMO

BACKGROUND: Clostridium difficile infection (CDI) incidence has increased dramatically over the last decade. Recent studies suggest that asymptomatic carriers may be an important reservoir of C. difficile in healthcare settings. We sought to identify the prevalence and risk factors for asymptomatic C. difficile carriage on admission to the hospital. METHODS: Patients admitted to Barnes-Jewish Hospital without diarrhea were enrolled from June 2010 through October 2011. Demographic information and healthcare and medication exposures 90 days prior to admission were collected. Stool specimens or rectal swabs were collected within 48 hours of admission and stored at -30°C until cultured. Clostridium difficile isolates were typed and compared with isolates from patients with CDI. RESULTS: A stool/swab specimen was obtained for 259 enrolled subjects on admission. Two hundred four (79%) were not colonized, 40 (15%) had toxigenic C. difficile (TCD), and 15 (6%) had nontoxigenic C. difficile. There were no differences between TCD-colonized and -uncolonized subjects for age (mean, 56 vs 58 years; P = .46), comorbidities, admission from another healthcare facility (33% vs 24%; P = .23), or recent hospitalization (50% vs 50%; P = .43). There were no differences in antimicrobial exposures in the 90 days prior to admission (55% vs 56%; P = .91). Asymptomatic carriers were colonized with strains similar to strains from patients with CDI, but the relative proportions were different. CONCLUSIONS: There was a high prevalence of TCD colonization on admission. In contrast to past studies, TCD colonization was not associated with recent antimicrobial or healthcare exposures. Additional investigation is needed to determine the role of asymptomatic TCD carriers on hospital-onset CDI incidence.


Assuntos
Portador Sadio/epidemiologia , Clostridioides difficile/isolamento & purificação , Infecções por Clostridium/epidemiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Portador Sadio/microbiologia , Clostridioides difficile/classificação , Clostridioides difficile/genética , Infecções por Clostridium/microbiologia , Fezes/microbiologia , Humanos , Pessoa de Meia-Idade , Prevalência , Estudos Prospectivos , Reto/microbiologia , Fatores de Risco , Arábia Saudita , Adulto Jovem
3.
Cell Rep ; 39(2): 110649, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35417701

RESUMO

Antibiotics are deployed against bacterial pathogens, but their targeting of conserved microbial processes means they also collaterally perturb the commensal microbiome. To understand acute and persistent effects of antibiotics on the gut microbiota of healthy adult volunteers, we quantify microbiome dynamics before, during, and 6 months after exposure to 4 commonly used antibiotic regimens. We observe an acute decrease in species richness and culturable bacteria after antibiotics, with most healthy adult microbiomes returning to pre-treatment species richness after 2 months, but with an altered taxonomy, resistome, and metabolic output, as well as an increased antibiotic resistance burden. Azithromycin delays the recovery of species richness, resulting in greater compositional distance. A subset of volunteers experience a persistent reduction in microbiome diversity after antibiotics and share compositional similarities with patients hospitalized in intensive care units. These results improve our quantitative understanding of the impact of antibiotics on commensal microbiome dynamics, resilience, and recovery.


Assuntos
Microbioma Gastrointestinal , Microbiota , Adulto , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bactérias , Humanos , Simbiose
4.
Infect Control Hosp Epidemiol ; 43(2): 167-173, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-33820576

RESUMO

OBJECTIVE: Alteration of the colonic microbiota following antimicrobial exposure allows colonization by antimicrobial-resistant organisms (AROs). Ingestion of a probiotic, such as Lactobacillus rhamnosus GG (LGG), could prevent colonization or infection with AROs by promoting healthy colonic microbiota. The purpose of this trial was to determine the effect of LGG administration on ARO colonization in hospitalized patients receiving antibiotics. DESIGN: Prospective, double-blinded, randomized controlled trial of LGG versus placebo among patients receiving broad-spectrum antibiotics. SETTING: Tertiary care center. PATIENTS: In total, 88 inpatients receiving broad-spectrum antibiotics were enrolled. INTERVENTION: Patients were randomized to receive 1 capsule containing 1×1010 cells of LGG twice daily (n = 44) or placebo (n = 44), stratified by ward type. Stool or rectal-swab specimens were collected for culture at enrollment, during admission, and at discharge. Using selective media, specimens were cultured for Clostridioides difficile, vancomycin-resistant Enterococcus spp (VRE), and antibiotic-resistant gram-negative bacteria. The primary outcome was any ARO acquisition. Secondary outcomes included loss of any ARO if colonized at enrollment, and acquisition or loss of individual ARO. RESULTS: ARO colonization prevalence at study enrollment was similar (LGG 39% vs placebo 39%). We detected no difference in any ARO acquisition (LGG 30% vs placebo 33%; OR,1.19; 95% CI, 0.38-3.75) nor for any individual ARO acquisition. There was no difference in the loss of any ARO (LGG 18% vs placebo 24%; OR, 1.44; 95% CI, 0.27-7.68) nor for any individual ARO. CONCLUSION: LGG administration neither prevented acquisition of ARO nor accelerated loss of ARO colonization.


Assuntos
Lacticaseibacillus rhamnosus , Probióticos , Enterococos Resistentes à Vancomicina , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Humanos , Probióticos/uso terapêutico , Estudos Prospectivos
5.
Cell Host Microbe ; 30(7): 1034-1047.e6, 2022 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-35545083

RESUMO

Large-scale genomic studies have identified within-host adaptation as a hallmark of bacterial infections. However, the impact of physiological, metabolic, and immunological differences between distinct niches on the pathoadaptation of opportunistic pathogens remains elusive. Here, we profile the within-host adaptation and evolutionary trajectories of 976 isolates representing 119 lineages of uropathogenic Escherichia coli (UPEC) sampled longitudinally from both the gastrointestinal and urinary tracts of 123 patients with urinary tract infections. We show that lineages persisting in both niches within a patient exhibit increased allelic diversity. Habitat-specific selection results in niche-specific adaptive mutations and genes, putatively mediating fitness in either environment. Within-lineage inter-habitat genomic plasticity mediated by mobile genetic elements (MGEs) provides the opportunistic pathogen with a mechanism to adapt to the physiological conditions of either habitat, and reduced MGE richness is associated with recurrence in gut-adapted UPEC lineages. Collectively, our results establish niche-specific adaptation as a driver of UPEC within-host evolution.


Assuntos
Infecções por Escherichia coli , Proteínas de Escherichia coli , Adaptação ao Hospedeiro , Infecções Urinárias , Escherichia coli Uropatogênica , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Adaptação ao Hospedeiro/genética , Humanos , Sequências Repetitivas Dispersas , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica/genética
6.
mSphere ; 6(1)2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33441409

RESUMO

Clostridioides difficile infection (CDI) is most commonly diagnosed using nucleic acid amplification tests (NAAT); the low positive predictive value of these assays results in patients colonized with C. difficile unnecessarily receiving CDI treatment antibiotics. The risks and benefits of antibiotic treatment in individuals with such cases are unknown. Fecal samples of NAAT-positive, toxin enzyme immunoassay (EIA)-negative patients were collected before, during, and after randomization to vancomycin (n = 8) or placebo (n = 7). C. difficile and antibiotic-resistant organisms (AROs) were selectively cultured from fecal and environmental samples. Shotgun metagenomics and comparative isolate genomics were used to understand the impact of oral vancomycin on the microbiome and environmental contamination. Overall, 80% of placebo patients and 71% of vancomycin patients were colonized with C. difficile posttreatment. One person randomized to placebo subsequently received treatment for CDI. In the vancomycin-treated group, beta-diversity (P = 0.0059) and macrolide-lincosamide-streptogramin (MLS) resistance genes (P = 0.037) increased after treatment; C. difficile and vancomycin-resistant enterococci (VRE) environmental contamination was found in 53% of patients and 26% of patients, respectively. We found that vancomycin alters the gut microbiota, does not permanently clear C. difficile, and is associated with VRE colonization/environmental contamination. (This study has been registered at ClinicalTrials.gov under registration no. NCT03388268.)IMPORTANCE A gold standard diagnostic for Clostridioides difficile infection (CDI) does not exist. An area of controversy is how to manage patients whose stool tests positive by nucleic acid amplification tests but negative by toxin enzyme immunoassay. Existing data suggest most of these patients do not have CDI, but most are treated with oral vancomycin. Potential benefits to treatment include a decreased risk for adverse outcomes if the patient does have CDI and the potential to decrease C. difficile shedding/transmission. However, oral vancomycin perturbs the intestinal microbiota and promotes antibiotic-resistant organism colonization/transmission. We conducted a double-blinded randomized controlled trial to assess the risk-benefit of oral vancomycin treatment in this population. Oral vancomycin did not result in long-term clearance of C. difficile, perturbed the microbiota, and was associated with colonization/shedding of vancomycin-resistant enterococci. This work underscores the need to better understand this population of patients in the context of C. difficile/ARO-related outcomes and transmission.


Assuntos
Antibacterianos/administração & dosagem , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/tratamento farmacológico , Fezes/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Vancomicina/administração & dosagem , Administração Oral , Adulto , Idoso , Antibacterianos/efeitos adversos , Antibacterianos/uso terapêutico , Clostridioides difficile/fisiologia , Feminino , Microbioma Gastrointestinal/genética , Humanos , Masculino , Metagenômica/métodos , Pessoa de Meia-Idade , Vancomicina/efeitos adversos , Vancomicina/uso terapêutico , Enterococos Resistentes à Vancomicina/efeitos dos fármacos , Enterococos Resistentes à Vancomicina/genética , Enterococos Resistentes à Vancomicina/isolamento & purificação
7.
Infect Control Hosp Epidemiol ; 41(5): 539-546, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31969206

RESUMO

OBJECTIVE: To assess potential transmission of antibiotic-resistant organisms (AROs) using surrogate markers and bacterial cultures. DESIGN: Pilot study. SETTING: A 1,260-bed tertiary-care academic medical center. PARTICIPANTS: The study included 25 patients (17 of whom were on contact precautions for AROs) and 77 healthcare personnel (HCP). METHODS: Fluorescent powder (FP) and MS2 bacteriophage were applied in patient rooms. HCP visits to each room were observed for 2-4 hours; hand hygiene (HH) compliance was recorded. Surfaces inside and outside the room and HCP skin and clothing were assessed for fluorescence, and swabs were collected for MS2 detection by polymerase chain reaction (PCR) and selective bacterial cultures. RESULTS: Transfer of FP was observed for 20 rooms (80%) and 26 HCP (34%). Transfer of MS2 was detected for 10 rooms (40%) and 15 HCP (19%). Bacterial cultures were positive for 1 room and 8 HCP (10%). Interactions with patients on contact precautions resulted in fewer FP detections than interactions with patients not on precautions (P < .001); MS2 detections did not differ by patient isolation status. Fluorescent powder detections did not differ by HCP type, but MS2 was recovered more frequently from physicians than from nurses (P = .03). Overall, HH compliance was better among HCP caring for patients on contact precautions than among HCP caring for patients not on precautions (P = .003), among nurses than among other nonphysician HCP at room entry (P = .002), and among nurses than among physicians at room exit (P = .03). Moreover, HCP who performed HH prior to assessment had fewer fluorescence detections (P = .008). CONCLUSIONS: Contact precautions were associated with greater HCP HH compliance and reduced detection of FP and MS2.


Assuntos
Infecção Hospitalar/microbiologia , Infecção Hospitalar/transmissão , Contaminação de Equipamentos , Centros Médicos Acadêmicos , Bacteriófagos/isolamento & purificação , Farmacorresistência Bacteriana Múltipla , Fidelidade a Diretrizes , Higiene das Mãos , Pessoal de Saúde , Hospitalização , Hospitais , Humanos , Pacientes , Projetos Piloto
8.
mBio ; 10(4)2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31455657

RESUMO

The rise of antimicrobial resistance in uropathogens has complicated the management of urinary tract infections (UTIs), particularly in patients who are afflicted by recurrent episodes of UTIs. Antimicrobial-resistant (AR) uropathogens persistently colonizing individuals at asymptomatic time points have been implicated in the pathophysiology of UTIs. The dynamics of uropathogen persistence following the resolution of symptomatic disease are, however, mostly unclear. To further our understanding, we determined longitudinal AR uropathogen carriage and clonal persistence of uropathogenic Escherichia coli, Proteus mirabilis, and Klebsiella pneumoniae isolates in the intestinal and urinary tracts of patients affected by recurrent and nonrecurrent UTIs. Clonal tracking of isolates in consecutively collected urine and fecal specimens indicated repeated transmission of uropathogens between the urinary tract and their intestinal reservoir. Our results further implicate three independent routes of recurrence of UTIs: (i) following an intestinal bloom of uropathogenic bacteria and subsequent bladder colonization, (ii) reinfection of the urinary tract from an external source, and (iii) bacterial persistence within the urinary tract. Taken together, our observation of clonal persistence following UTIs and uropathogen transmission between the intestinal and urinary tracts warrants further investigations into the connection between the intestinal microbiome and recurrent UTIs.IMPORTANCE The increasing antimicrobial resistance of uropathogens is challenging the continued efficacy of empiric antibiotic therapy for UTIs, which are among the most frequent bacterial infections worldwide. It has been suggested that drug-resistant uropathogens could persist in the intestine after the resolution of UTI and cause recurrences following periurethral contamination. A better understanding of the transmission dynamics between the intestinal and urinary tracts, combined with phenotypic characterization of the uropathogen populations in both habitats, could inform prudent therapies designed to overcome the rising resistance of uropathogens. Here, we integrate genomic surveillance with clinical microbiology to show that drug-resistant clones persist within and are readily transmitted between the intestinal and urinary tracts of patients affected by recurrent and nonrecurrent UTIs. Thus, our results advocate for understanding persistent intestinal uropathogen colonization as part of the pathophysiology of UTIs, particularly in patients affected by recurrent episodes of symptomatic disease.


Assuntos
Antibacterianos/farmacologia , Escherichia coli/genética , Genômica , Klebsiella pneumoniae/genética , Proteus mirabilis/genética , Infecções Urinárias/microbiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Escherichia coli/efeitos dos fármacos , Humanos , Klebsiella pneumoniae/efeitos dos fármacos , Estudos Longitudinais , Pessoa de Meia-Idade , Filogenia , Proteus mirabilis/efeitos dos fármacos , Recidiva
9.
mSphere ; 3(6)2018 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-30463925

RESUMO

Strategies to prevent multidrug-resistant organism (MDRO) infections are scarce, but autologous fecal microbiota transplantation (autoFMT) may limit gastrointestinal MDRO expansion. AutoFMT involves banking one's feces during a healthy state for later use in restoring gut microbiota following perturbation. This pilot study evaluated the effect of autoFMT on gastrointestinal microbiome taxonomic composition, resistance gene content, and metabolic capacity after exposure to amoxicillin-clavulanic acid (Amox-Clav). Ten healthy participants were enrolled. All received 5 days of Amox-Clav. Half were randomized to autoFMT, derived from stool collected pre-antimicrobial exposure, by enema, and half to saline enema. Participants submitted stool samples pre- and post-Amox-Clav and enema and during a 90-day follow-up period. Shotgun metagenomic sequencing revealed taxonomic composition, resistance gene content, and metabolic capacity. Amox-Clav significantly altered gut taxonomic composition in all participants (n = 10, P < 0.01); however, only three participants exhibited major changes at the phylum level following exposure. In the cohort as a whole, beta-lactamase genes were enriched following Amox-Clav (P < 0.05), and predicted metabolic capacity was significantly altered (P < 0.01). Species composition, metabolic capacity, and beta-lactamase abundance returned to pre-antimicrobial exposure state 7 days after either autoFMT or saline enema (P > 0.05, compared to enrollment). Alterations to microbial metabolic capacity occurred following antimicrobial exposure even in participants without substantial taxonomic disruption, potentially creating open niches for pathogen colonization. Our findings suggest that metabolic potential is an important consideration for complete assessment of antimicrobial impact on the microbiome. AutoFMT was well tolerated and may have contributed to phylogenetic recovery. (This study has been registered at ClinicalTrials.gov under identifier NCT02046525.)IMPORTANCE The spread of multidrug resistance among pathogenic organisms threatens the efficacy of antimicrobial treatment options. The human gut serves as a reservoir for many drug-resistant organisms and their resistance genes, and perturbation of the gut microbiome by antimicrobial exposure can open metabolic niches to resistant pathogens. Once established in the gut, antimicrobial-resistant bacteria can persist even after antimicrobial exposure ceases. Strategies to prevent multidrug-resistant organism (MDRO) infections are scarce, but autologous fecal microbiota transplantation (autoFMT) may limit gastrointestinal MDRO expansion. AutoFMT involves banking one's feces during a healthy state for later use in restoring gut microbiota following perturbation. This pilot study evaluated the effect of amoxicillin-clavulanic acid (Amox-Clav) exposure and autoFMT on gastrointestinal microbiome taxonomic composition, resistance gene content, and metabolic capacity. Importantly, we found that metabolic capacity was perturbed even in cases where gross phylogeny remained unchanged and that autoFMT was safe and well tolerated.


Assuntos
Combinação Amoxicilina e Clavulanato de Potássio/administração & dosagem , Antibacterianos/administração & dosagem , Transplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal , Metabolismo , Microbiota , Inibidores de beta-Lactamases/administração & dosagem , Adulto , Enema , Feminino , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Adulto Jovem
10.
Transplant Direct ; 3(4): e145, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28405601

RESUMO

BACKGROUND: Clostridium difficile infection (CDI) is a frequent cause of diarrhea among allogeneic hematopoietic cell transplant (HCT) recipients. It is unknown whether risk factors for CDI vary by time posttransplant. METHODS: We performed a 3-year prospective cohort study of CDI in allogeneic HCT recipients. Participants were enrolled during their transplant hospitalizations. Clinical assessments were performed weekly during hospitalizations and for 12 weeks posttransplant, and monthly for 30 months thereafter. Data were collected through patient interviews and chart review, and included CDI diagnosis, demographics, transplant characteristics, medications, infections, and outcomes. CDI cases were included if they occurred within 1 year of HCT and were stratified by time from transplant. Multivariable logistic regression was used to determine risk factors for CDI. RESULTS: One hundred eighty-seven allogeneic HCT recipients were enrolled, including 63 (34%) patients who developed CDI. 38 (60%) CDI cases occurred during the preengraftment period (days 0-30 post-HCT) and 25 (40%) postengraftment (day >30). Lack of any preexisting comorbid disease was significantly associated with lower risk of CDI preengraftment (odds ratio [OR], 0.3; 95% confidence interval [CI], 0.1-0.9). Relapsed underlying disease (OR, 6.7; 95% CI, 1.3-33.1), receipt of any high-risk antimicrobials (OR, 11.8; 95% CI, 2.9-47.8), and graft-versus-host disease (OR, 7.8; 95% CI, 2.0-30.2) were significant independent risk factors for CDI postengraftment. CONCLUSIONS: A large portion of CDI cases occurred during the postengraftment period in allogeneic HCT recipients, suggesting that surveillance for CDI should continue beyond the transplant hospitalization and preengraftment period. Patients with continued high underlying severity of illness were at increased risk of CDI postengraftment.

11.
Infect Control Hosp Epidemiol ; 38(9): 1077-1083, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28606192

RESUMO

OBJECTIVE To evaluate healthcare worker (HCW) risk of self-contamination when donning and doffing personal protective equipment (PPE) using fluorescence and MS2 bacteriophage. DESIGN Prospective pilot study. SETTING Tertiary-care hospital. PARTICIPANTS A total of 36 HCWs were included in this study: 18 donned/doffed contact precaution (CP) PPE and 18 donned/doffed Ebola virus disease (EVD) PPE. INTERVENTIONS HCWs donned PPE according to standard protocols. Fluorescent liquid and MS2 bacteriophage were applied to HCWs. HCWs then doffed their PPE. After doffing, HCWs were scanned for fluorescence and swabbed for MS2. MS2 detection was performed using reverse transcriptase PCR. The donning and doffing processes were videotaped, and protocol deviations were recorded. RESULTS Overall, 27% of EVD PPE HCWs and 50% of CP PPE HCWs made ≥1 protocol deviation while donning, and 100% of EVD PPE HCWs and 67% of CP PPE HCWs made ≥1 protocol deviation while doffing (P=.02). The median number of doffing protocol deviations among EVD PPE HCWs was 4, versus 1 among CP PPE HCWs. Also, 15 EVD PPE protocol deviations were committed by doffing assistants and/or trained observers. Fluorescence was detected on 8 EVD PPE HCWs (44%) and 5 CP PPE HCWs (28%), most commonly on hands. MS2 was recovered from 2 EVD PPE HCWs (11%) and 3 CP PPE HCWs (17%). CONCLUSIONS Protocol deviations were common during both EVD and CP PPE doffing, and some deviations during EVD PPE doffing were committed by the HCW doffing assistant and/or the trained observer. Self-contamination was common. PPE donning/doffing are complex and deserve additional study. Infect Control Hosp Epidemiol 2017;38:1077-1083.


Assuntos
Fidelidade a Diretrizes/estatística & dados numéricos , Levivirus/isolamento & purificação , Roupa de Proteção/virologia , Dispositivos de Proteção Respiratória/virologia , Adulto , Feminino , Luvas Protetoras/virologia , Pessoal de Saúde , Humanos , Controle de Infecções/métodos , Controle de Infecções/normas , Masculino , Pessoa de Meia-Idade , Missouri , Equipamento de Proteção Individual/virologia , Projetos Piloto , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Centros de Atenção Terciária , Raios Ultravioleta , Gravação em Vídeo
12.
Infect Control Hosp Epidemiol ; 37(12): 1401-1407, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27691986

RESUMO

OBJECTIVE To determine whether Clostridium difficile is present in the food of hospitalized patients and to estimate the risk of subsequent colonization associated with C. difficile in food. METHODS This was a prospective cohort study of inpatients at a university-affiliated tertiary care center, May 9, 2011-July 12, 2012. Enrolled patients submitted a portion of food from each meal. Patient stool specimens and/or rectal swabs were collected at enrollment, every 3 days thereafter, and at discharge, and were cultured for C. difficile. Clinical data were reviewed for evidence of infection due to C. difficile. A stochastic, discrete event model was developed to predict exposure to C. difficile from food, and the estimated number of new colonization events from food exposures per 1,000 admissions was determined. RESULTS A total of 149 patients were enrolled and 910 food specimens were obtained. Two food specimens from 2 patients were positive for C. difficile (0.2% of food samples; 1.3% of patients). Neither of the 2 patients was colonized at baseline with C. difficile. Discharge colonization status was available for 1 of the 2 patients and was negative. Neither was diagnosed with C. difficile infection while hospitalized or during the year before or after study enrollment. Stochastic modeling indicated contaminated hospital food would be responsible for less than 1 newly colonized patient per 1,000 hospital admissions. CONCLUSIONS The recovery of C. difficile from the food of hospitalized patients was rare. Modeling suggests hospital food is unlikely to be a source of C. difficile acquisition. Infect Control Hosp Epidemiol 2016;1401-1407.


Assuntos
Clostridioides difficile/isolamento & purificação , Infecção Hospitalar/microbiologia , Microbiologia de Alimentos , Centros Médicos Acadêmicos , Adulto , Idoso , Idoso de 80 Anos ou mais , Enterocolite Pseudomembranosa , Fezes/microbiologia , Feminino , Hospitalização , Humanos , Pacientes Internados , Masculino , Pessoa de Meia-Idade , Missouri , Estudos Prospectivos , Adulto Jovem
14.
Chest ; 124(3): 1021-9, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12970033

RESUMO

OBJECTIVE: To determine the influence of microbiologically confirmed infection on hospital mortality among patients requiring intensive care for > 48 h. DESIGN: Prospective cohort study. SETTING: Medical ICU of the Barnes-Jewish Hospital, an urban teaching hospital. PATIENTS: A total of 893 patients requiring intensive care for > 48 h. INTERVENTIONS: Prospective patient surveillance and data collection. MEASUREMENTS AND MAIN RESULTS: Three hundred seventy-two patients (41.7%) requiring intensive care for > 48 h had a microbiologically confirmed infection. Only six patients (0.7% [1.6% of patients with microbiologically confirmed infections]) received inadequate antimicrobial therapy during the first 24 h of treatment, and 248 patients (27.8%) died during hospitalization. Compared to hospital survivors, hospital nonsurvivors were significantly more likely to have a microbiologically confirmed infection (53.2% vs 37.2%, respectively; p < 0.001) and to develop severe sepsis (45.6% vs 28.7%, respectively; p < 0.001). Cirrhosis and the requirement for vasopressors were the only variables identified by multiple logistic regression analysis as independent risk factors for hospital mortality in all patient groupings of severity of illness. Multiple logistic regression analysis also demonstrated that underlying malignancy (adjusted odds ratio [AOR], 1.98; 95% CI, 1.55 to 2.53), chronic renal insufficiency (AOR, 1.57; 95% CI, 1.31 to 1.87), cirrhosis (AOR, 1.94; 95% CI, 1.48 to 2.53), temperature > 38.3 degrees C (AOR, 1.72; 95% CI, 1.47 to 2.02), severe sepsis (AOR, 2.78; 95% CI, 1.94 to 3.98), positive culture for vancomycin-resistant enterococci (AOR, 1.78; 95% CI, 1.51 to 2.09), and the presence of multiple infections (AOR, 1.65; 95% CI, 1.28 to 2.14) were independently associated with the requirement for therapy with vasopressors. CONCLUSIONS: Microbiologically confirmed infections are common among patients requiring medical intensive care for > 48 h. Despite the administration of adequate antimicrobial therapy, microbiologically confirmed infections appear to be an important cause of hemodynamic instability and increased hospital mortality. These data suggest that clinical efforts aimed at the prevention of infections and improvements in the medical management of patients with severe infections, especially those associated with hemodynamic instability and the need for vasopressors, are required to achieve further improvements in patient outcomes.


Assuntos
Cuidados Críticos/estatística & dados numéricos , Infecção Hospitalar/mortalidade , Mortalidade Hospitalar , Choque Séptico/mortalidade , População Urbana/estatística & dados numéricos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Causas de Morte , Estudos de Coortes , Feminino , Humanos , Falência Renal Crônica/complicações , Falência Renal Crônica/mortalidade , Cirrose Hepática/complicações , Cirrose Hepática/mortalidade , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Missouri , Neoplasias/complicações , Neoplasias/mortalidade , Razão de Chances , Estudos Prospectivos , Risco , Taxa de Sobrevida , Vasoconstritores/administração & dosagem
15.
Infect Control Hosp Epidemiol ; 24(4): 257-63, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12725354

RESUMO

OBJECTIVE: To determine the epidemiology of colonization with vancomycin-resistant Enterococcus (VRE) among intensive care unit (ICU) patients. DESIGN: Ten-month prospective cohort study. SETTING: A 19-bed medical ICU of a 1,440-bed teaching hospital. METHODS: Patients admitted to the ICU had rectal swab cultures for VRE on admission and weekly thereafter. VRE-positive patients were cared for using contact precautions. Clinical data, including microbiology reports, were collected prospectively during the ICU stay. RESULTS: Of 519 patients who had admission stool cultures, 127 (25%) had cultures that were positive for VRE. Risk factors for VRE colonization identified by multiple logistic regression analysis were hospital stay greater than 3 days prior to ICU admission (adjusted odds ratio [AOR], 3.6; 95% confidence interval [CI95], 2.3 to 5.7), chronic dialysis (AOR, 2.4; CI95, 1.2 to 4.5), and having been admitted to the study hospital one to two times (AOR, 2.3; CI95, 1.4 to 3.8) or more than two times (AOR, 6.5; CI95, 3.7 to 11.6) within the past 12 months. Of the 352 VRE-negative patients who had one or more follow-up cultures, 74 (21%) became VRE positive during their ICU stay (27 cases per 1,000 patient-ICU days). CONCLUSION: The prevalence of VRE culture positivity on ICU admission was high and a sizable fraction of ICU patients became VRE positive during their ICU stay despite contact precautions for VRE-positive patients. This was likely due in large part to prior VRE exposures in the rest of the hospital where these control measures were not being used.


Assuntos
Enterococcus/efeitos dos fármacos , Enterococcus/patogenicidade , Unidades de Terapia Intensiva , Resistência a Vancomicina , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Infecção Hospitalar , Enterococcus/isolamento & purificação , Feminino , Hospitais de Ensino/estatística & dados numéricos , Humanos , Controle de Infecções , Masculino , Pessoa de Meia-Idade , Prevalência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA