Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 565(7741): 654-658, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30675060

RESUMEN

Diffuse gliomas are the most common malignant brain tumours in adults and include glioblastomas and World Health Organization (WHO) grade II and grade III tumours (sometimes referred to as lower-grade gliomas). Genetic tumour profiling is used to classify disease and guide therapy1,2, but involves brain surgery for tissue collection; repeated tumour biopsies may be necessary for accurate genotyping over the course of the disease3-10. While the detection of circulating tumour DNA (ctDNA) in the blood of patients with primary brain tumours remains challenging11,12, sequencing of ctDNA from the cerebrospinal fluid (CSF) may provide an alternative way to genotype gliomas with lower morbidity and cost13,14. We therefore evaluated the representation of the glioma genome in CSF from 85 patients with gliomas who underwent a lumbar puncture because they showed neurological signs or symptoms. Here we show that tumour-derived DNA was detected in CSF from 42 out of 85 patients (49.4%) and was associated with disease burden and adverse outcome. The genomic landscape of glioma in the CSF included a broad spectrum of genetic alterations and closely resembled the genomes of tumour biopsies. Alterations that occur early during tumorigenesis, such as co-deletion of chromosome arms 1p and 19q (1p/19q codeletion) and mutations in the metabolic genes isocitrate dehydrogenase 1 (IDH1) or IDH21,2, were shared in all matched ctDNA-positive CSF-tumour pairs, whereas growth factor receptor signalling pathways showed considerable evolution. The ability to monitor the evolution of the glioma genome through a minimally invasive technique could advance the clinical development and use of genotype-directed therapies for glioma, one of the most aggressive human cancers.


Asunto(s)
Evolución Molecular , Glioma/líquido cefalorraquídeo , Glioma/genética , Biopsia Líquida , Mutación , Genes Relacionados con las Neoplasias/genética , Genoma Humano/genética , Genómica , Glioblastoma/líquido cefalorraquídeo , Glioblastoma/genética , Glioblastoma/patología , Glioma/patología , Humanos , Clasificación del Tumor
2.
Nature ; 517(7533): 205-8, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25337874

RESUMEN

The gastrointestinal tracts of mammals are colonized by hundreds of microbial species that contribute to health, including colonization resistance against intestinal pathogens. Many antibiotics destroy intestinal microbial communities and increase susceptibility to intestinal pathogens. Among these, Clostridium difficile, a major cause of antibiotic-induced diarrhoea, greatly increases morbidity and mortality in hospitalized patients. Which intestinal bacteria provide resistance to C. difficile infection and their in vivo inhibitory mechanisms remain unclear. Here we correlate loss of specific bacterial taxa with development of infection, by treating mice with different antibiotics that result in distinct microbiota changes and lead to varied susceptibility to C. difficile. Mathematical modelling augmented by analyses of the microbiota of hospitalized patients identifies resistance-associated bacteria common to mice and humans. Using these platforms, we determine that Clostridium scindens, a bile acid 7α-dehydroxylating intestinal bacterium, is associated with resistance to C. difficile infection and, upon administration, enhances resistance to infection in a secondary bile acid dependent fashion. Using a workflow involving mouse models, clinical studies, metagenomic analyses, and mathematical modelling, we identify a probiotic candidate that corrects a clinically relevant microbiome deficiency. These findings have implications for the rational design of targeted antimicrobials as well as microbiome-based diagnostics and therapeutics for individuals at risk of C. difficile infection.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Clostridioides difficile/fisiología , Susceptibilidad a Enfermedades/microbiología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Microbiota/fisiología , Animales , Antibacterianos/farmacología , Evolución Biológica , Clostridioides difficile/efectos de los fármacos , Clostridium/metabolismo , Colitis/metabolismo , Colitis/microbiología , Colitis/prevención & control , Colitis/terapia , Heces/microbiología , Femenino , Humanos , Intestinos/efectos de los fármacos , Metagenoma/genética , Ratones , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Microbiota/genética , Simbiosis
3.
Blood ; 131(26): 2978-2986, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29674425

RESUMEN

Respiratory viral infections are frequent in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HCT) and can potentially progress to lower respiratory tract infection (LRTI). The intestinal microbiota contributes to resistance against viral and bacterial pathogens in the lung. However, whether intestinal microbiota composition and associated changes in microbe-derived metabolites contribute to the risk of LRTI following upper respiratory tract viral infection remains unexplored in the setting of allo-HCT. Fecal samples from 360 allo-HCT patients were collected at the time of stem cell engraftment and subjected to deep, 16S ribosomal RNA gene sequencing to determine microbiota composition, and short-chain fatty acid levels were determined in a nested subset of fecal samples. The development of respiratory viral infections and LRTI was determined for 180 days following allo-HCT. Clinical and microbiota risk factors for LRTI were subsequently evaluated using survival analysis. Respiratory viral infection occurred in 149 (41.4%) patients. Of those, 47 (31.5%) developed LRTI. Patients with higher abundances of butyrate-producing bacteria were fivefold less likely to develop viral LRTI, independent of other factors (adjusted hazard ratio = 0.22, 95% confidence interval 0.04-0.69). Higher representation of butyrate-producing bacteria in the fecal microbiota is associated with increased resistance against respiratory viral infection with LRTI in allo-HCT patients.


Asunto(s)
Bacterias/metabolismo , Butiratos/metabolismo , Microbioma Gastrointestinal , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Infecciones del Sistema Respiratorio/etiología , Infecciones del Sistema Respiratorio/microbiología , Virosis/etiología , Virosis/microbiología , Adulto , Heces/microbiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores Protectores , Infecciones del Sistema Respiratorio/metabolismo , Trasplante Homólogo/efectos adversos , Virosis/metabolismo
4.
Am J Transplant ; 19(2): 488-500, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29920927

RESUMEN

Posttransplant diarrhea is associated with kidney allograft failure and death, but its etiology remains unknown in the majority of cases. Because altered gut microbial ecology is a potential basis for diarrhea, we investigated whether posttransplant diarrhea is associated with gut dysbiosis. We enrolled 71 kidney allograft recipients for serial fecal specimen collections in the first 3 months of transplantation and profiled the gut microbiota using 16S ribosomal RNA (rRNA) gene V4-V5 deep sequencing. The Shannon diversity index was significantly lower in 28 diarrheal fecal specimens from 25 recipients with posttransplant diarrhea than in 112 fecal specimens from 46 recipients without posttransplant diarrhea. We found a lower relative abundance of 13 commensal genera (Benjamini-Hochberg adjusted P ≤ .15) in the diarrheal fecal specimens including the same 4 genera identified in our prior study. The 28 diarrheal fecal specimens were also evaluated by a multiplexed polymerase chain reaction (PCR) assay for 22 bacterial, viral, and protozoan gastrointestinal pathogens, and 26 specimens were negative for infectious etiologies. Using PICRUSt (Phylogenetic Investigation of Communities by Reconstruction of Unobserved States) to predict metagenomic functions, we found that diarrheal fecal specimens had a lower abundance of metabolic genes. Our findings suggest that posttransplant diarrhea is not associated with common infectious diarrheal pathogens but with a gut dysbiosis.


Asunto(s)
Bacterias/crecimiento & desarrollo , Diarrea/etiología , Disbiosis/etiología , Microbioma Gastrointestinal , Rechazo de Injerto/etiología , Fallo Renal Crónico/cirugía , Trasplante de Riñón/efectos adversos , Adulto , Bacterias/genética , Bacterias/aislamiento & purificación , Estudios de Casos y Controles , Estudios de Cohortes , Diarrea/patología , Disbiosis/patología , Heces/microbiología , Femenino , Estudios de Seguimiento , Tasa de Filtración Glomerular , Rechazo de Injerto/patología , Supervivencia de Injerto , Humanos , Pruebas de Función Renal , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias , Pronóstico , ARN Ribosómico 16S/genética , Factores de Riesgo
5.
J Infect Dis ; 215(7): 1117-1123, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28498996

RESUMEN

Background: Clostridium difficile infection (CDI) is a frequent complication in recipients of allogeneic hematopoietic stem cell transplantation (allo-HSCT), who receive intensive treatments that significantly disrupt the intestinal microbiota. In this study, we examined the microbiota composition of allo-HSCT recipients to identify bacterial colonizers that confer protection against CDI after engraftment. Methods: Feces collected from adult recipients allo-HSCT at engraftment were analyzed; 16S ribosomal RNA genes were sequenced and analyzed from each sample. Bacterial taxa with protective effects against development of CDI were identified by means of linear discriminant analysis effect size analysis and then further assessed with clinical predictors of CDI using survival analysis. Results: A total of 234 allo-HSCT recipients were studied; postengraftment CDI developed in 53 (22.6%). Within the composition of the microbiota, the presence of 3 distinct bacterial taxa was correlated with protection against CDI: Bacteroidetes, Lachnospiraceae, and Ruminococcaceae. Colonization with these groups at engraftment was associated with a 60% lower risk of CDI, independent of clinical factors. Conclusions: Colonization with these 3 bacterial groups is associated with a lower risk of CDI. These groups have been shown to be vital components of the intestinal microbiota. Targeted efforts to maintain them may help minimize the risk of CDI in this at-risk population.


Asunto(s)
Infecciones por Clostridium/microbiología , Microbioma Gastrointestinal , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Adulto , Bacteroidetes/clasificación , Bacteroidetes/aislamiento & purificación , Clostridiales/clasificación , Clostridiales/aislamiento & purificación , Clostridioides difficile , Heces/microbiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores Protectores , ARN Ribosómico 16S/genética , Trasplante Homólogo
6.
Cancer Causes Control ; 28(9): 959-969, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28762074

RESUMEN

PURPOSE: Poor oral health appears to be a risk factor for pancreatic cancer, possibly implicating the oral microbiota. In this pilot study, we evaluated the characteristics of the oral microbiota in patients with pancreatic ductal adenocarcinoma (PDAC), intraductal papillary mucinous neoplasms (IPMN), and healthy controls. METHODS: Forty newly diagnosed PDAC patients, 39 IPMN patients, and 58 controls, excluding current smokers and users of antibiotics, provided saliva samples. Common oral bacterial species were comprehensively surveyed by sequencing of the 16S rRNA microbial genes. We obtained measures of diversity and the mean relative proportions of individual taxa. We explored the degree to which these measures differed according to respondent characteristics based on individual interviews. RESULTS: PDAC cases did not differ in diversity measures from either controls or IPMN cases. PDAC cases had higher mean relative proportions of Firmicutes and related taxa, while controls had higher mean relative proportions of Proteobacteria and related taxa. Results were generally similar when comparing PDAC to IPMN cases. Among IPMNs and controls combined, younger individuals had higher levels of several taxa within the Proteobacteria. The only other variable consistently related to mean relative proportions was mouthwash use, with taxa within Firmicutes more common among users. CONCLUSIONS: While there were no differences in diversity of the oral microbiota among these groups, there were differences in the mean relative proportions of some taxa. Characteristics of the oral microbiota are not associated with most measures of oral health.


Asunto(s)
Bacterias/aislamiento & purificación , Carcinoma Ductal Pancreático/microbiología , Microbiota , Boca/microbiología , Neoplasias Pancreáticas/microbiología , Anciano , Bacterias/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , ARN Ribosómico 16S/genética
7.
PLoS Pathog ; 11(9): e1005132, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26334306

RESUMEN

Antibiotic resistance among enterococci and γ-proteobacteria is an increasing problem in healthcare settings. Dense colonization of the gut by antibiotic-resistant bacteria facilitates their spread between patients and also leads to bloodstream and other systemic infections. Antibiotic-mediated destruction of the intestinal microbiota and consequent loss of colonization resistance are critical factors leading to persistence and spread of antibiotic-resistant bacteria. The mechanisms underlying microbiota-mediated colonization resistance remain incompletely defined and are likely distinct for different antibiotic-resistant bacterial species. It is unclear whether enterococci or γ-proteobacteria, upon expanding to high density in the gut, confer colonization resistance against competing bacterial species. Herein, we demonstrate that dense intestinal colonization with vancomycin-resistant Enterococcus faecium (VRE) does not reduce in vivo growth of carbapenem-resistant Klebsiella pneumoniae. Reciprocally, K. pneumoniae does not impair intestinal colonization by VRE. In contrast, transplantation of a diverse fecal microbiota eliminates both VRE and K. pneumoniae from the gut. Fluorescence in situ hybridization demonstrates that VRE and K. pneumoniae localize to the same regions in the colon but differ with respect to stimulation and invasion of the colonic mucus layer. While VRE and K. pneumoniae occupy the same three-dimensional space within the gut lumen, their independent growth and persistence in the gut suggests that they reside in distinct niches that satisfy their specific in vivo metabolic needs.


Asunto(s)
Enteritis/microbiología , Enterococcus faecium/fisiología , Infecciones por Bacterias Grampositivas/microbiología , Mucosa Intestinal/microbiología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/fisiología , Enterococos Resistentes a la Vancomicina/fisiología , Ampicilina/efectos adversos , Animales , Antibacterianos/efectos adversos , Antibacterianos/farmacología , Carbapenémicos/farmacología , Recuento de Colonia Microbiana , Farmacorresistencia Bacteriana , Enteritis/patología , Enteritis/prevención & control , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/crecimiento & desarrollo , Enterococcus faecium/aislamiento & purificación , Trasplante de Microbiota Fecal , Heces/microbiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Infecciones por Bacterias Grampositivas/patología , Infecciones por Bacterias Grampositivas/prevención & control , Interacciones Huésped-Patógeno , Hibridación Fluorescente in Situ , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Infecciones por Klebsiella/patología , Infecciones por Klebsiella/prevención & control , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/crecimiento & desarrollo , Klebsiella pneumoniae/aislamiento & purificación , Ratones Endogámicos C57BL , Interacciones Microbianas , Organismos Libres de Patógenos Específicos , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/crecimiento & desarrollo , Enterococos Resistentes a la Vancomicina/aislamiento & purificación
8.
Blood ; 124(7): 1174-82, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-24939656

RESUMEN

Highly diverse bacterial populations inhabit the gastrointestinal tract and modulate host inflammation and promote immune tolerance. In allogeneic hematopoietic stem cell transplantation (allo-HSCT), the gastrointestinal mucosa is damaged, and colonizing bacteria are impacted, leading to an impaired intestinal microbiota with reduced diversity. We examined the impact of intestinal diversity on subsequent mortality outcomes following transplantation. Fecal specimens were collected from 80 recipients of allo-HSCT at the time of stem cell engraftment. Bacterial 16S rRNA gene sequences were characterized, and microbial diversity was estimated using the inverse Simpson index. Subjects were classified into high, intermediate, and low diversity groups and assessed for differences in outcomes. Mortality outcomes were significantly worse in patients with lower intestinal diversity; overall survival at 3 years was 36%, 60%, and 67% for low, intermediate, and high diversity groups, respectively (P = .019, log-rank test). Low diversity showed a strong effect on mortality after multivariate adjustment for other clinical predictors (transplant related mortality: adjusted hazard ratio, 5.25; P = .014). In conclusion, the diversity of the intestinal microbiota at engraftment is an independent predictor of mortality in allo-HSCT recipients. These results indicate that the intestinal microbiota may be an important factor in the success or failure in allo-HSCT.


Asunto(s)
Tracto Gastrointestinal/microbiología , Variación Genética , Trasplante de Células Madre Hematopoyéticas/métodos , Microbiota/genética , Adulto , Bacterias/clasificación , Bacterias/genética , Heces/microbiología , Femenino , Trasplante de Células Madre Hematopoyéticas/mortalidad , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Análisis Multivariante , Evaluación de Resultado en la Atención de Salud/métodos , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , Filogenia , Pronóstico , Modelos de Riesgos Proporcionales , ARN Ribosómico 16S/genética , Tasa de Supervivencia , Trasplante Homólogo
9.
J Infect Dis ; 212(10): 1656-65, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25920320

RESUMEN

Antibiotic administration disrupts the intestinal microbiota, increasing susceptibility to pathogens such as Clostridium difficile. Metronidazole or oral vancomycin can cure C. difficile infection, and administration of these agents to prevent C. difficile infection in high-risk patients, although not sanctioned by Infectious Disease Society of America guidelines, has been considered. The relative impacts of metronidazole and vancomycin on the intestinal microbiota and colonization resistance are unknown. We investigated the effect of brief treatment with metronidazole and/or oral vancomycin on susceptibility to C. difficile, vancomycin-resistant Enterococcus, carbapenem-resistant Klebsiella pneumoniae, and Escherichia coli infection in mice. Although metronidazole resulted in transient loss of colonization resistance, oral vancomycin markedly disrupted the microbiota, leading to prolonged loss of colonization resistance to C. difficile infection and dense colonization by vancomycin-resistant Enterococcus, K. pneumoniae, and E. coli. Our results demonstrate that vancomycin, and to a lesser extent metronidazole, are associated with marked intestinal microbiota destruction and greater risk of colonization by nosocomial pathogens.


Asunto(s)
Antiinfecciosos/administración & dosificación , Infecciones Bacterianas/inmunología , Resistencia a la Enfermedad/efectos de los fármacos , Metronidazol/administración & dosificación , Vancomicina/administración & dosificación , Animales , Antiinfecciosos/efectos adversos , Infecciones Bacterianas/microbiología , Clostridioides difficile/aislamiento & purificación , Modelos Animales de Enfermedad , Escherichia coli/aislamiento & purificación , Femenino , Klebsiella pneumoniae/aislamiento & purificación , Metronidazol/efectos adversos , Ratones Endogámicos C57BL , Vancomicina/efectos adversos , Enterococos Resistentes a la Vancomicina/aislamiento & purificación
10.
Biol Blood Marrow Transplant ; 21(8): 1373-83, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25977230

RESUMEN

The relationship between intestinal microbiota composition and acute graft-versus-host disease (GVHD) after allogeneic blood/marrow transplantation (allo-BMT) is not well understood. Intestinal bacteria have long been thought to contribute to GVHD pathophysiology, but recent animal studies in nontransplant settings have found that anti-inflammatory effects are mediated by certain subpopulations of intestinal commensals. Hypothesizing that a more nuanced relationship may exist between the intestinal bacteria and GVHD, we evaluated the fecal bacterial composition of 64 patients 12 days after BMT. We found that increased bacterial diversity was associated with reduced GVHD-related mortality. Furthermore, harboring increased amounts of bacteria belonging to the genus Blautia was associated with reduced GVHD lethality in this cohort and was confirmed in another independent cohort of 51 patients from the same institution. Blautia abundance was also associated with improved overall survival. We evaluated the abundance of Blautia with respect to clinical factors and found that loss of Blautia was associated with treatment with antibiotics that inhibit anaerobic bacteria and receiving total parenteral nutrition for longer durations. We conclude that increased abundance of commensal bacteria belonging to the Blautia genus is associated with reduced lethal GVHD and improved overall survival.


Asunto(s)
Bacterias/metabolismo , Enfermedad Injerto contra Huésped/mortalidad , Intestinos/microbiología , Estudios de Cohortes , Femenino , Humanos , Mucosa Intestinal/metabolismo , Masculino , Factores de Riesgo , Análisis de Supervivencia
11.
Infect Immun ; 81(3): 965-73, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23319552

RESUMEN

Bacteria causing infections in hospitalized patients are increasingly antibiotic resistant. Classical infection control practices are only partially effective at preventing spread of antibiotic-resistant bacteria within hospitals. Because the density of intestinal colonization by the highly antibiotic-resistant bacterium vancomycin-resistant Enterococcus (VRE) can exceed 10(9) organisms per gram of feces, even optimally implemented hygiene protocols often fail. Decreasing the density of intestinal colonization, therefore, represents an important approach to limit VRE transmission. We demonstrate that reintroduction of a diverse intestinal microbiota to densely VRE-colonized mice eliminates VRE from the intestinal tract. While oxygen-tolerant members of the microbiota are ineffective at eliminating VRE, administration of obligate anaerobic commensal bacteria to mice results in a billionfold reduction in the density of intestinal VRE colonization. 16S rRNA gene sequence analysis of intestinal bacterial populations isolated from mice that cleared VRE following microbiota reconstitution revealed that recolonization with a microbiota that contains Barnesiella correlates with VRE elimination. Characterization of the fecal microbiota of patients undergoing allogeneic hematopoietic stem cell transplantation demonstrated that intestinal colonization with Barnesiella confers resistance to intestinal domination and bloodstream infection with VRE. Our studies indicate that obligate anaerobic bacteria belonging to the Barnesiella genus enable clearance of intestinal VRE colonization and may provide novel approaches to prevent the spread of highly antibiotic-resistant bacteria.


Asunto(s)
Bacteroidaceae/fisiología , Enterococcus faecium/efectos de los fármacos , Infecciones por Bacterias Grampositivas/terapia , Intestinos/microbiología , Resistencia a la Vancomicina , Animales , ADN Bacteriano , Femenino , Ratones , Ratones Endogámicos C57BL , ARN Bacteriano/genética , ARN Ribosómico 16S/genética
12.
Nat Commun ; 13(1): 3477, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35710741

RESUMEN

PD-1 blockade (nivolumab) efficacy remains modest for metastatic sarcoma. In this paper, we present an open-label, non-randomized, non-comparative pilot study of bempegaldesleukin, a CD122-preferential interleukin-2 pathway agonist, with nivolumab in refractory sarcoma at Memorial Sloan Kettering/MD Anderson Cancer Centers (NCT03282344). We report on the primary outcome of objective response rate (ORR) and secondary endpoints of toxicity, clinical benefit, progression-free survival, overall survival, and durations of response/treatment. In 84 patients in 9 histotype cohorts, all patients experienced ≥1 adverse event and treatment-related adverse event; 1 death was possibly treatment-related. ORR was highest in angiosarcoma (3/8) and undifferentiated pleomorphic sarcoma (2/10), meeting predefined endpoints. Results of our exploratory investigation of predictive biomarkers show: CD8 + T cell infiltrates and PD-1 expression correlate with improved ORR; upregulation of immune-related pathways correlate with improved efficacy; Hedgehog pathway expression correlate with resistance. Exploration of this combination in selected sarcomas, and of Hedgehog signaling as a predictive biomarker, warrants further study in larger cohorts.


Asunto(s)
Antineoplásicos Inmunológicos , Neoplasias Primarias Secundarias , Sarcoma , Antineoplásicos Inmunológicos/uso terapéutico , Proteínas Hedgehog , Humanos , Interleucina-2/uso terapéutico , Neoplasias Primarias Secundarias/inducido químicamente , Nivolumab/uso terapéutico , Proyectos Piloto , Receptor de Muerte Celular Programada 1/metabolismo , Sarcoma/tratamiento farmacológico , Sarcoma/patología
13.
Artículo en Inglés | MEDLINE | ID: mdl-33163850

RESUMEN

PURPOSE: Although primary germ cell tumors (GCTs) have been extensively characterized, molecular analysis of metastatic sites has been limited. We performed whole-exome sequencing and targeted next-generation sequencing on paired primary and metastatic GCT samples in a patient cohort enriched for cisplatin-resistant disease. PATIENTS AND METHODS: Tissue sequencing was performed on 100 tumor specimens from 50 patients with metastatic GCT, and sequencing of plasma cell-free DNA was performed for a subset of patients. RESULTS: The mutational landscape of primary and metastatic pairs from GCT patients was highly discordant (68% of all somatic mutations were discordant). Whereas genome duplication was common and highly concordant between primary and metastatic samples, only 25% of primary-metastasis pairs had ≥ 50% concordance at the level of DNA copy number alterations (CNAs). Evolutionary-based analyses revealed that most mutations arose after CNAs at the respective loci in both primary and metastatic samples, with oncogenic mutations enriched in the set of early-occurring mutations versus variants of unknown significance (VUSs). TP53 pathway alterations were identified in nine cisplatin-resistant patients and had the highest degree of concordance in primary and metastatic specimens, consistent with their association with this treatment-resistant phenotype. CONCLUSION: Analysis of paired primary and metastatic GCT specimens revealed significant molecular heterogeneity for both CNAs and somatic mutations. Among loci demonstrating serial genetic evolution, most somatic mutations arose after CNAs, but oncogenic mutations were enriched in the set of early-occurring mutations as compared with VUSs. Alterations in TP53 were clonal when present and shared among primary-metastasis pairs.

14.
mBio ; 10(2)2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862751

RESUMEN

A diverse, antibiotic-naive microbiota prevents highly antibiotic-resistant microbes, including carbapenem-resistant Klebsiella pneumoniae (CR-Kp), from achieving dense colonization of the intestinal lumen. Antibiotic-mediated destruction of the microbiota leads to expansion of CR-Kp in the gut, markedly increasing the risk of bacteremia in vulnerable patients. While preventing dense colonization represents a rational approach to reduce intra- and interpatient dissemination of CR-Kp, little is known about pathogen-associated factors that enable dense growth and persistence in the intestinal lumen. To identify genetic factors essential for dense colonization of the gut by CR-Kp, we constructed a highly saturated transposon mutant library with >150,000 unique mutations in an ST258 strain of CR-Kp and screened for in vitro growth and in vivo intestinal colonization in antibiotic-treated mice. Stochastic and partially reversible fluctuations in the representation of different mutations during dense colonization revealed the dynamic nature of intestinal microbial populations. We identified genes that are crucial for early and late stages of dense gut colonization and confirmed their role by testing isogenic mutants in in vivo competition assays with wild-type CR-Kp Screening of the transposon library also identified mutations that enhanced in vivo CR-Kp growth. These newly identified colonization factors may provide novel therapeutic opportunities to reduce intestinal colonization by CR-KpIMPORTANCEKlebsiella pneumoniae is a common cause of bloodstream infections in immunocompromised and hospitalized patients, and over the last 2 decades, some strains have acquired resistance to nearly all available antibiotics, including broad-spectrum carbapenems. The U.S. Centers for Disease Control and Prevention has listed carbapenem-resistant K. pneumoniae (CR-Kp) as an urgent public health threat. Dense colonization of the intestine by CR-Kp and other antibiotic-resistant bacteria is associated with an increased risk of bacteremia. Reducing the density of gut colonization by CR-Kp is likely to reduce their transmission from patient to patient in health care facilities as well as systemic infections. How CR-Kp expands and persists in the gut lumen, however, is poorly understood. Herein, we generated a highly saturated mutant library in a multidrug-resistant K. pneumoniae strain and identified genetic factors that are associated with dense gut colonization by K. pneumoniae This study sheds light on host colonization by K. pneumoniae and identifies potential colonization factors that contribute to high-density persistence of K. pneumoniae in the intestine.


Asunto(s)
Antibacterianos/administración & dosificación , Carbapenémicos/administración & dosificación , Genoma Bacteriano , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/crecimiento & desarrollo , Factores de Virulencia/genética , Resistencia betalactámica , Animales , Antibacterianos/farmacología , Carbapenémicos/farmacología , Elementos Transponibles de ADN , Modelos Animales de Enfermedad , Pruebas Genéticas , Klebsiella pneumoniae/genética , Ratones , Mutagénesis Insercional
15.
Nat Commun ; 10(1): 5521, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31797927

RESUMEN

The origin of most bacterial infections in the urinary tract is often presumed to be the gut. Herein, we investigate the relationship between the gut microbiota and future development of bacteriuria and urinary tract infection (UTI). We perform gut microbial profiling using 16S rRNA gene deep sequencing on 510 fecal specimens from 168 kidney transplant recipients and metagenomic sequencing on a subset of fecal specimens and urine supernatant specimens. We report that a 1% relative gut abundance of Escherichia is an independent risk factor for Escherichia bacteriuria and UTI and a 1% relative gut abundance of Enterococcus is an independent risk factor for Enterococcus bacteriuria. Strain analysis establishes a close strain level alignment between species found in the gut and in the urine in the same subjects. Our results support a gut microbiota-UTI axis, suggesting that modulating the gut microbiota may be a potential novel strategy to prevent UTIs.


Asunto(s)
Bacterias/genética , Infecciones Bacterianas/microbiología , ADN Bacteriano/genética , ARN Ribosómico 16S/genética , Infecciones Urinarias/microbiología , Bacterias/clasificación , Bacteriuria/etiología , Bacteriuria/microbiología , Bacteriuria/orina , ADN Bacteriano/análisis , Infecciones por Escherichia coli/etiología , Infecciones por Escherichia coli/microbiología , Heces/microbiología , Microbioma Gastrointestinal/genética , Humanos , Trasplante de Riñón/efectos adversos , Trasplante de Riñón/métodos , Factores de Riesgo , Infecciones Urinarias/etiología , Infecciones Urinarias/orina
16.
Blood Adv ; 3(13): 2040-2044, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31289031

RESUMEN

Patients with multiple myeloma (MM) who achieve minimal residual disease (MRD) negativity after upfront treatment have superior outcomes compared with those who remain MRD+ Recently, associations have been shown between specific commensal microbes and development of plasma cell disorders. Here, we report the association between intestinal microbiota composition and treatment outcome in MM. Microbiota composition of fecal samples collected from 34 MM patients after induction therapy and at the time of flow cytometry-based bone marrow MRD testing was determined by 16S ribosomal RNA sequencing. We observed a higher relative abundance of Eubacterium hallii in the 16 MRD- patients relative to the 18 MRD+ patients. No association was observed between microbial relative abundance and autologous stem cell transplantation history or MM paraprotein isotype. No differences in microbiota α diversity were observed between MRD- and MRD+ patients. The potential association of microbiota composition with treatment response in MM patients is an important parameter for additional correlative and clinical investigation.


Asunto(s)
Microbioma Gastrointestinal , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/etiología , Neoplasia Residual/patología , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Médula Ósea/patología , Terapia Combinada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/terapia , Estadificación de Neoplasias , Resultado del Tratamiento
17.
Int J Nanomedicine ; 13: 5275-5286, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30237714

RESUMEN

BACKGROUND: Amine-modified carbon nanotubes are drug delivery platforms with great potential that have not yet been applied in human clinical trials. Although modified nanotube vectors have the ability to carry multiple effectors, targeting agents, and even wrapped RNA, reports on unmodified, insoluble carbon nanotubes have highlighted inflammation in organs, including the intestine, with disruption of its resident microbiota. Disruption of the microbiota may allow for colonization by pathogenic bacteria, such as Clostridoidies difficile, stimulate immunoinfiltrates into the lamina propria or alter the absorption of therapeutics. Most proposed nanotube drugs are soluble, modified structures that are administered parenterally, and the majority of these soluble macromolecules are renally excreted; however, some are released into the bile, gaining access to the gastrointestinal tract. METHODS: Using environmentally isolated BALB/C mice in oral and intraperitoneal dosing models, high dose (3.80 or 4.25 mg/week), we administered amine-modified, soluble carbon nanotubes for 7 or 8 weeks. The general health and weight of the mice were monitored weekly, and upon killing, the diversity and content of their colonic, cecal, and ileal microbiota were assessed using shotgun 16S DNA sequencing. RESULTS AND CONCLUSION: We show that while oral administration at suprapharmacological doses modestly altered the α- and ß-diversity of the mouse microbiome, these changes did not result in observed changes in clinical end points. Intraperitoneally-dosed mice exhibited none of the toxicities assessed.


Asunto(s)
Aminas/química , Microbiota , Nanotubos de Carbono/química , Administración Oral , Animales , Bacterias/metabolismo , Biodiversidad , Peso Corporal , Femenino , Tracto Gastrointestinal/microbiología , Humanos , Lisina/química , Ratones Endogámicos BALB C , Nanotubos de Carbono/toxicidad , Análisis de Componente Principal
18.
Sci Transl Med ; 10(460)2018 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-30257956

RESUMEN

Antibiotic treatment can deplete the commensal bacteria of a patient's gut microbiota and, paradoxically, increase their risk of subsequent infections. In allogeneic hematopoietic stem cell transplantation (allo-HSCT), antibiotic administration is essential for optimal clinical outcomes but significantly disrupts intestinal microbiota diversity, leading to loss of many beneficial microbes. Although gut microbiota diversity loss during allo-HSCT is associated with increased mortality, approaches to reestablish depleted commensal bacteria have yet to be developed. We have initiated a randomized, controlled clinical trial of autologous fecal microbiota transplantation (auto-FMT) versus no intervention and have analyzed the intestinal microbiota profiles of 25 allo-HSCT patients (14 who received auto-FMT treatment and 11 control patients who did not). Changes in gut microbiota diversity and composition revealed that the auto-FMT intervention boosted microbial diversity and reestablished the intestinal microbiota composition that the patient had before antibiotic treatment and allo-HSCT. These results demonstrate the potential for fecal sample banking and posttreatment remediation of a patient's gut microbiota after microbiota-depleting antibiotic treatment during allo-HSCT.


Asunto(s)
Antibacterianos/farmacología , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Biodiversidad , Microbioma Gastrointestinal/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Humanos , Estudios Longitudinales , Trasplante Autólogo
19.
mBio ; 8(4)2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28790208

RESUMEN

Clostridium difficile is a spore-forming anaerobic bacterium that causes colitis in patients with disrupted colonic microbiota. While some individuals are asymptomatic C. difficile carriers, symptomatic disease ranges from mild diarrhea to potentially lethal toxic megacolon. The wide disease spectrum has been attributed to the infected host's age, underlying diseases, immune status, and microbiome composition. However, strain-specific differences in C. difficile virulence have also been implicated in determining colitis severity. Because patients infected with C. difficile are unique in terms of medical history, microbiome composition, and immune competence, determining the relative contribution of C. difficile virulence to disease severity has been challenging, and conclusions regarding the virulence of specific strains have been inconsistent. To address this, we used a mouse model to test 33 clinical C. difficile strains isolated from patients with disease severities ranging from asymptomatic carriage to severe colitis, and we determined their relative in vivo virulence in genetically identical, antibiotic-pretreated mice. We found that murine infections with C. difficile clade 2 strains (including multilocus sequence type 1/ribotype 027) were associated with higher lethality and that C. difficile strains associated with greater human disease severity caused more severe disease in mice. While toxin production was not strongly correlated with in vivo colonic pathology, the ability of C. difficile strains to grow in the presence of secondary bile acids was associated with greater disease severity. Whole-genome sequencing and identification of core and accessory genes identified a subset of accessory genes that distinguish high-virulence from lower-virulence C. difficile strains.IMPORTANCEClostridium difficile is an important cause of hospital-associated intestinal infections, and recent years have seen an increase in the number and severity of cases in the United States. A patient's antibiotic history, immune status, and medical comorbidities determine, in part, the severity of C. difficile infection. The relative virulence of different clinical C. difficile strains, although postulated to determine disease severity in patients, has been more difficult to consistently associate with mild versus severe colitis. We tested 33 distinct clinical C. difficile isolates for their ability to cause disease in genetically identical mice and found that C. difficile strains belonging to clade 2 were associated with higher mortality. Differences in survival were not attributed to differences in toxin production but likely resulted from the distinct gene content in the various clinical isolates.


Asunto(s)
Clostridioides difficile/genética , Clostridioides difficile/patogenicidad , Genoma Bacteriano , Factores de Virulencia/genética , Animales , Infecciones Asintomáticas , Toxinas Bacterianas , Ácidos y Sales Biliares/farmacología , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/crecimiento & desarrollo , Infecciones por Clostridium/microbiología , Colitis/microbiología , Infección Hospitalaria , Diarrea/microbiología , Modelos Animales de Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones
20.
Cell Host Microbe ; 21(5): 592-602.e4, 2017 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-28494240

RESUMEN

Antibiotic-mediated microbiota destruction and the consequent loss of colonization resistance can result in intestinal domination with vancomycin-resistant Enterococcus (VRE), leading to bloodstream infection in hospitalized patients. Clearance of VRE remains a challenging goal that, if achieved, would reduce systemic VRE infections and patient-to-patient transmission. Although obligate anaerobic commensal bacteria have been associated with colonization resistance to VRE, the specific bacterial species involved remain undefined. Herein, we demonstrate that a precisely defined consortium of commensal bacteria containing the Clostridium cluster XIVa species Blautia producta and Clostridium bolteae restores colonization resistance against VRE and clears VRE from the intestines of mice. While C. bolteae did not directly mediate VRE clearance, it enabled intestinal colonization with B. producta, which directly inhibited VRE growth. These findings suggest that therapeutic or prophylactic administration of defined bacterial consortia to individuals with compromised microbiota composition may reduce inter-patient transmission and intra-patient dissemination of highly antibiotic-resistant pathogens.


Asunto(s)
Enterococcus faecium/crecimiento & desarrollo , Microbiota/fisiología , Simbiosis/fisiología , Enterococos Resistentes a la Vancomicina/crecimiento & desarrollo , Ampicilina/farmacología , Animales , Antibacterianos/farmacología , Bacterias/genética , Bacterias/aislamiento & purificación , Fenómenos Fisiológicos Bacterianos , Clostridium/fisiología , Recuento de Colonia Microbiana , ADN Bacteriano , Farmacorresistencia Bacteriana , Enterococcus faecium/patogenicidad , Heces/microbiología , Femenino , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/prevención & control , Intestinos/microbiología , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética , Vancomicina/farmacología , Enterococos Resistentes a la Vancomicina/genética , Enterococos Resistentes a la Vancomicina/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA