Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Infect Control Hosp Epidemiol ; 44(6): 997-1001, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35591782

RESUMEN

Data from the National Healthcare Safety Network were analyzed to assess the impact of COVID-19 on the incidence of healthcare-associated infections (HAI) during 2021. Standardized infection ratios were significantly higher than those during the prepandemic period, particularly during 2021-Q1 and 2021-Q3. The incidence of HAI was elevated during periods of high COVID-19 hospitalizations.


Asunto(s)
COVID-19 , Infección Hospitalaria , Humanos , COVID-19/epidemiología , Incidencia , Pandemias , Infección Hospitalaria/epidemiología , Atención a la Salud
3.
Infect Control Hosp Epidemiol ; 43(6): 790-793, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-33719981

RESUMEN

Data reported to the Centers for Disease Control and Prevention's National Healthcare Safety Network (CDC NHSN) were analyzed to understand the potential impact of the COVID-19 pandemic on central-line-associated bloodstream infections (CLABSIs) in acute-care hospitals. Descriptive analysis of the standardized infection ratio (SIR) was conducted by location, location type, geographic area, and bed size.


Asunto(s)
COVID-19 , Infecciones Relacionadas con Catéteres , Infección Hospitalaria , Sepsis , COVID-19/epidemiología , Infecciones Relacionadas con Catéteres/epidemiología , Infección Hospitalaria/epidemiología , Atención a la Salud , Humanos , Pandemias , Sepsis/epidemiología
4.
Infect Control Hosp Epidemiol ; 43(1): 12-25, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34473013

RESUMEN

OBJECTIVES: To determine the impact of the coronavirus disease 2019 (COVID-19) pandemic on healthcare-associated infection (HAI) incidence in US hospitals, national- and state-level standardized infection ratios (SIRs) were calculated for each quarter in 2020 and compared to those from 2019. METHODS: Central-line-associated bloodstream infections (CLABSIs), catheter-associated urinary tract infections (CAUTIs), ventilator-associated events (VAEs), select surgical site infections, and Clostridioides difficile and methicillin-resistant Staphylococcus aureus (MRSA) bacteremia laboratory-identified events reported to the National Healthcare Safety Network for 2019 and 2020 by acute-care hospitals were analyzed. SIRs were calculated for each HAI and quarter by dividing the number of reported infections by the number of predicted infections, calculated using 2015 national baseline data. Percentage changes between 2019 and 2020 SIRs were calculated. Supporting analyses, such as an assessment of device utilization in 2020 compared to 2019, were also performed. RESULTS: Significant increases in the national SIRs for CLABSI, CAUTI, VAE, and MRSA bacteremia were observed in 2020. Changes in the SIR varied by quarter and state. The largest increase was observed for CLABSI, and significant increases in VAE incidence and ventilator utilization were seen across all 4 quarters of 2020. CONCLUSIONS: This report provides a national view of the increases in HAI incidence in 2020. These data highlight the need to return to conventional infection prevention and control practices and build resiliency in these programs to withstand future pandemics.


Asunto(s)
COVID-19 , Infecciones Relacionadas con Catéteres , Infección Hospitalaria , Staphylococcus aureus Resistente a Meticilina , Neumonía Asociada al Ventilador , Infecciones Relacionadas con Catéteres/epidemiología , Infección Hospitalaria/epidemiología , Infección Hospitalaria/prevención & control , Atención a la Salud , Humanos , Neumonía Asociada al Ventilador/epidemiología , Neumonía Asociada al Ventilador/prevención & control , SARS-CoV-2
5.
JMIR Public Health Surveill ; 7(7): e23528, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34328436

RESUMEN

BACKGROUND: The Centers for Disease Control and Prevention's (CDC's) National Healthcare Safety Network (NHSN) is the most widely used health care-associated infection (HAI) and antimicrobial use and resistance surveillance program in the United States. Over 37,000 health care facilities participate in the program and submit a large volume of surveillance data. These data are used by the facilities themselves, the CDC, and other agencies and organizations for a variety of purposes, including infection prevention, antimicrobial stewardship, and clinical quality measurement. Among the summary metrics made available by the NHSN are standardized infection ratios, which are used to identify HAI prevention needs and measure progress at the national, regional, state, and local levels. OBJECTIVE: To extend the use of geospatial methods and tools to NHSN data, and in turn to promote and inspire new uses of the rendered data for analysis and prevention purposes, we developed a web-enabled system that enables integrated visualization of HAI metrics and supporting data. METHODS: We leveraged geocoding and visualization technologies that are readily available and in current use to develop a web-enabled system designed to support visualization and interpretation of data submitted to the NHSN from geographically dispersed sites. The server-client model-based system enables users to access the application via a web browser. RESULTS: We integrated multiple data sets into a single-page dashboard designed to enable users to navigate across different HAI event types, choose specific health care facility or geographic locations for data displays, and scale across time units within identified periods. We launched the system for internal CDC use in January 2019. CONCLUSIONS: CDC NHSN statisticians, data analysts, and subject matter experts identified opportunities to extend the use of geospatial methods and tools to NHSN data and provided the impetus to develop NHSNViz. The development effort proceeded iteratively, with the developer adding or enhancing functionality and including additional data sets in a series of prototype versions, each of which incorporated user feedback. The initial production version of NHSNViz provides a new geospatial analytic resource built in accordance with CDC user requirements and extensible to additional users and uses in subsequent versions.


Asunto(s)
Infección Hospitalaria , Centers for Disease Control and Prevention, U.S. , Infección Hospitalaria/epidemiología , Infección Hospitalaria/prevención & control , Atención a la Salud , Instituciones de Salud , Humanos , Estados Unidos/epidemiología
6.
Nat Med ; 24(10): 1499-1503, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30275568

RESUMEN

We report a patient relapsing 9 months after CD19-targeted CAR T cell (CTL019) infusion with CD19- leukemia that aberrantly expressed the anti-CD19 CAR. The CAR gene was unintentionally introduced into a single leukemic B cell during T cell manufacturing, and its product bound in cis to the CD19 epitope on the surface of leukemic cells, masking it from recognition by and conferring resistance to CTL019.


Asunto(s)
Antígenos CD19/inmunología , Resistencia a Antineoplásicos/inmunología , Epítopos/inmunología , Leucemia/tratamiento farmacológico , Adulto , Antígenos CD19/uso terapéutico , Linfocitos B/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Leucemia/inmunología , Leucemia/patología , Masculino , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/uso terapéutico , Receptores Quiméricos de Antígenos , Linfocitos T/inmunología , Adulto Joven
7.
Cancer Immunol Res ; 6(9): 1100-1109, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30030295

RESUMEN

The success of chimeric antigen receptor (CAR)-mediated immunotherapy in acute lymphoblastic leukemia (ALL) highlights the potential of T-cell therapies with directed cytotoxicity against specific tumor antigens. The efficacy of CAR T-cell therapy depends on the engraftment and persistence of T cells following adoptive transfer. Most protocols for T-cell engineering routinely expand T cells ex vivo for 9 to 14 days. Because the potential for engraftment and persistence is related to the state of T-cell differentiation, we hypothesized that reducing the duration of ex vivo culture would limit differentiation and enhance the efficacy of CAR T-cell therapy. We demonstrated that T cells with a CAR-targeting CD19 (CART19) exhibited less differentiation and enhanced effector function in vitro when harvested from cultures at earlier (day 3 or 5) compared with later (day 9) timepoints. We then compared the therapeutic potential of early versus late harvested CART19 in a murine xenograft model of ALL and showed that the antileukemic activity inversely correlated with ex vivo culture time: day 3 harvested cells showed robust tumor control despite using a 6-fold lower dose of CART19, whereas day 9 cells failed to control leukemia at limited cell doses. We also demonstrated the feasibility of an abbreviated culture in a large-scale current good manufacturing practice-compliant process. Limiting the interval between T-cell isolation and CAR treatment is critical for patients with rapidly progressing disease. Generating CAR T cells in less time also improves potency, which is central to the effectiveness of these therapies. Cancer Immunol Res; 6(9); 1100-9. ©2018 AACR.


Asunto(s)
Técnicas de Cultivo de Célula , Inmunoterapia Adoptiva , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Animales , Antígenos CD19/genética , Antígenos CD19/inmunología , Antígenos de Neoplasias/inmunología , Diferenciación Celular/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Humanos , Activación de Linfocitos , Ratones , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
JCI Insight ; 3(1)2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29321369

RESUMEN

Successful tumor eradication by chimeric antigen receptor-expressing (CAR-expressing) T lymphocytes depends on CAR T cell persistence and effector function. We hypothesized that CD4+ and CD8+ T cells may exhibit distinct persistence and effector phenotypes, depending on the identity of specific intracellular signaling domains (ICDs) used to generate the CAR. First, we demonstrate that the ICOS ICD dramatically enhanced the in vivo persistence of CAR-expressing CD4+ T cells that, in turn, increased the persistence of CD8+ T cells expressing either CD28- or 4-1BB-based CARs. These data indicate that persistence of CD8+ T cells was highly dependent on a helper effect provided by the ICD used to redirect CD4+ T cells. Second, we discovered that combining ICOS and 4-1BB ICDs in a third-generation CAR displayed superior antitumor effects and increased persistence in vivo. Interestingly, we found that the membrane-proximal ICD displayed a dominant effect over the distal domain in third-generation CARs. The optimal antitumor and persistence benefits observed in third-generation ICOSBBz CAR T cells required the ICOS ICD to be positioned proximal to the cell membrane and linked to the ICOS transmembrane domain. Thus, CARs with ICOS and 4-1BB ICD demonstrate increased efficacy in solid tumor models over our current 4-1BB-based CAR and are promising therapeutics for clinical testing.


Asunto(s)
Ligando 4-1BB/metabolismo , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Adenocarcinoma , Animales , Antineoplásicos/farmacología , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos , Línea Celular Tumoral , Membrana Celular , Humanos , Neoplasias Pulmonares , Ratones , Metástasis de la Neoplasia , Neoplasias Pancreáticas , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
9.
Immunity ; 44(2): 380-90, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26885860

RESUMEN

Chimeric antigen receptors (CARs) redirect T cell cytotoxicity against cancer cells, providing a promising approach to cancer immunotherapy. Despite extensive clinical use, the attributes of CAR co-stimulatory domains that impact persistence and resistance to exhaustion of CAR-T cells remain largely undefined. Here, we report the influence of signaling domains of coreceptors CD28 and 4-1BB on the metabolic characteristics of human CAR T cells. Inclusion of 4-1BB in the CAR architecture promoted the outgrowth of CD8(+) central memory T cells that had significantly enhanced respiratory capacity, increased fatty acid oxidation and enhanced mitochondrial biogenesis. In contrast, CAR T cells with CD28 domains yielded effector memory cells with a genetic signature consistent with enhanced glycolysis. These results provide, at least in part, a mechanistic insight into the differential persistence of CAR-T cells expressing 4-1BB or CD28 signaling domains in clinical trials and inform the design of future CAR T cell therapies.


Asunto(s)
Antígenos CD28/metabolismo , Linfocitos T CD8-positivos/fisiología , Vacunas contra el Cáncer/inmunología , Inmunoterapia , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Antígenos CD28/genética , Respiración de la Célula , Células Cultivadas , Glucólisis , Humanos , Memoria Inmunológica , Metabolismo de los Lípidos , Mitocondrias/metabolismo , Neoplasias/inmunología , Receptor Cross-Talk , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/genética , Transducción de Señal/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
10.
Blood ; 127(9): 1117-27, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26813675

RESUMEN

Anti-CD19 chimeric antigen receptor (CAR) T-cell therapy is highly promising but requires robust T-cell expansion and engraftment. A T-cell defect in chronic lymphocytic leukemia (CLL) due to disease and/or therapy impairs ex vivo expansion and response to CAR T cells. To evaluate the effect of ibrutinib treatment on the T-cell compartment in CLL as it relates to CAR T-cell generation, we examined the phenotype and function of T cells in a cohort of CLL patients during their course of treatment with ibrutinib. We found that ≥5 cycles of ibrutinib therapy improved the expansion of CD19-directed CAR T cells (CTL019), in association with decreased expression of the immunosuppressive molecule programmed cell death 1 on T cells and of CD200 on B-CLL cells. In support of these findings, we observed that 3 CLL patients who had been treated with ibrutinib for ≥1 year at the time of T-cell collection had improved ex vivo and in vivo CTL019 expansion, which correlated positively together and with clinical response. Lastly, we show that ibrutinib exposure does not impair CAR T-cell function in vitro but does improve CAR T-cell engraftment, tumor clearance, and survival in human xenograft models of resistant acute lymphocytic leukemia and CLL when administered concurrently. Our collective findings indicate that ibrutinib enhances CAR T-cell function and suggest that clinical trials with combination therapy are warranted. Our studies demonstrate that improved T-cell function may also contribute to the efficacy of ibrutinib in CLL. These trials were registered at www.clinicaltrials.gov as #NCT01747486, #NCT01105247, and #NCT01217749.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/inmunología , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Adenina/análogos & derivados , Administración Oral , Anciano , Animales , Antígenos CD/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , Demografía , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Técnicas de Transferencia de Gen , Humanos , Terapia de Inmunosupresión , Células K562 , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Ratones , Persona de Mediana Edad , Piperidinas , Receptor de Muerte Celular Programada 1/metabolismo , Pirazoles/administración & dosificación , Pirazoles/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Linfocitos T/efectos de los fármacos , Factores de Tiempo , Resultado del Tratamiento
12.
Sci Transl Med ; 7(275): 275ra22, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25696001

RESUMEN

Chimeric antigen receptors (CARs) are synthetic molecules designed to redirect T cells to specific antigens. CAR-modified T cells can mediate long-term durable remissions in B cell malignancies, but expanding this platform to solid tumors requires the discovery of surface targets with limited expression in normal tissues. The variant III mutation of the epidermal growth factor receptor (EGFRvIII) results from an in-frame deletion of a portion of the extracellular domain, creating a neoepitope. We chose a vector backbone encoding a second-generation CAR based on efficacy of a murine scFv-based CAR in a xenograft model of glioblastoma. Next, we generated a panel of humanized scFvs and tested their specificity and function as soluble proteins and in the form of CAR-transduced T cells; a low-affinity scFv was selected on the basis of its specificity for EGFRvIII over wild-type EGFR. The lead candidate scFv was tested in vitro for its ability to direct CAR-transduced T cells to specifically lyse, proliferate, and secrete cytokines in response to antigen-bearing targets. We further evaluated the specificity of the lead CAR candidate in vitro against EGFR-expressing keratinocytes and in vivo in a model of mice grafted with normal human skin. EGFRvIII-directed CAR T cells were also able to control tumor growth in xenogeneic subcutaneous and orthotopic models of human EGFRvIII(+) glioblastoma. On the basis of these results, we have designed a phase 1 clinical study of CAR T cells transduced with humanized scFv directed to EGFRvIII in patients with either residual or recurrent glioblastoma (NCT02209376).


Asunto(s)
Neoplasias Encefálicas/terapia , Receptores ErbB/inmunología , Glioblastoma/terapia , Inmunoterapia , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Ratones
13.
Nanotoxicology ; 9(6): 737-48, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25378273

RESUMEN

The increasing risk of incidental exposure to nanomaterials has led to mounting concerns regarding nanotoxicity. Zinc oxide nanoparticles (ZnO NPs) are produced in large quantities and have come under scrutiny due to their capacity to cause cytotoxicity in vitro and potential to cause harm in vivo. Recent evidence has indicated that ZnO NPs promote autophagy in cells; however, the signaling pathways and the role of ion release inducing toxicity remain unclear. In this study, we report that ZnO NPs are immunotoxic to primary and immortalized immune cells. Importantly, such immunotoxicity is observed in mice in vivo, since death of splenocytes is seen after intranasal exposure to ZnO NPs. We determined that ZnO NPs release free Zn(2+) that can be taken up by immune cells, resulting in cell death. Inhibiting free Zn(2+) ions in solution with EDTA or their uptake with CaCl2 abrogates ZnO NP-induced cell death. ZnO NP-mediated immune cell death was associated with increased levels of intracellular reactive oxygen species (ROS). ZnO NP death was not due to apoptosis, necroptosis or pyroptosis. Exposure of immune cells to ZnO NPs resulted in autophagic death and increased levels of LC3A, an essential component of autophagic vacuoles. Accordingly, ZnO NP-mediated upregulation of LC3A and induction of immune cell death were inhibited by blocking autophagy and ROS production. We conclude that release of Zn(2+) from ZnO NPs triggers the production of excessive intracellular ROS, resulting in autophagic death of immune cells. Our findings suggest that exposure to ZnO NPs has the potential to impact host immunity.


Asunto(s)
Autofagia/efectos de los fármacos , Nanopartículas/toxicidad , Bazo/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Óxido de Zinc/toxicidad , Animales , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Citometría de Flujo , Humanos , Células Jurkat , Ratones Endogámicos C57BL , Nanopartículas/química , Tamaño de la Partícula , Especies Reactivas de Oxígeno/metabolismo , Bazo/inmunología , Propiedades de Superficie , Linfocitos T/metabolismo , Linfocitos T/patología , Óxido de Zinc/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...