Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
NPJ Biofilms Microbiomes ; 7(1): 20, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33658514

ABSTRACT

Little is known about the relationship between gut dysbiosis, inflammation, and adverse outcomes in patients with chronic kidney disease. We examined the association of microbial diversity with all-cause mortality in hemodialysis patients. The gut microbiota was assessed by 16S ribosomal RNA gene sequencing. During a median follow-up of 2.1 years, the adjusted risk of death among patients with higher diversity (above median) was 74% lower than that among patients with lower diversity (below median). We then compared the microbial composition between nonsurvivors and survivors in a matched case-control study. We observed significantly lower microbial diversity and higher proinflammatory cytokines among nonsurvivors than survivors. Specifically, the relative abundance of Succinivibrio and Anaerostipes, two short-chain fatty acid-producing bacteria, was markedly reduced in nonsurvivors. Thus, a unique gut microbial composition is associated with an increased risk of mortality among hemodialysis patients and may be used to identify subjects with a poor prognosis.


Subject(s)
Bacteria/classification , Dysbiosis/epidemiology , RNA, Ribosomal, 16S/genetics , Renal Dialysis/mortality , Renal Insufficiency, Chronic/microbiology , Aged , Aged, 80 and over , Bacteria/genetics , Bacteria/isolation & purification , Case-Control Studies , Dysbiosis/mortality , Feces/microbiology , Female , Gastrointestinal Microbiome , Humans , Male , Middle Aged , Phylogeny , Prognosis , Renal Insufficiency, Chronic/mortality , Sequence Analysis, DNA
2.
Am J Respir Crit Care Med ; 202(12): 1666-1677, 2020 12 15.
Article in English | MEDLINE | ID: mdl-32717152

ABSTRACT

Rationale: Host inflammatory responses have been strongly associated with adverse outcomes in critically ill patients, but the biologic underpinnings of such heterogeneous responses have not been defined.Objectives: We examined whether respiratory tract microbiome profiles are associated with host inflammation and clinical outcomes of acute respiratory failure.Methods: We collected oral swabs, endotracheal aspirates (ETAs), and plasma samples from mechanically ventilated patients. We performed 16S ribosomal RNA gene sequencing to characterize upper and lower respiratory tract microbiota and classified patients into host-response subphenotypes on the basis of clinical variables and plasma biomarkers of innate immunity and inflammation. We derived diversity metrics and composition clusters with Dirichlet multinomial models and examined our data for associations with subphenotypes and clinical outcomes.Measurements and Main Results: Oral and ETA microbial communities from 301 mechanically ventilated subjects had substantial heterogeneity in α and ß diversity. Dirichlet multinomial models revealed a cluster with low α diversity and enrichment for pathogens (e.g., high Staphylococcus or Pseudomonadaceae relative abundance) in 35% of ETA samples, associated with a hyperinflammatory subphenotype, worse 30-day survival, and longer time to liberation from mechanical ventilation (adjusted P < 0.05), compared with patients with higher α diversity and relative abundance of typical oral microbiota. Patients with evidence of dysbiosis (low α diversity and low relative abundance of "protective" oral-origin commensal bacteria) in both oral and ETA samples (17%, combined dysbiosis) had significantly worse 30-day survival and longer time to liberation from mechanical ventilation than patients without dysbiosis (55%; adjusted P < 0.05).Conclusions: Respiratory tract dysbiosis may represent an important, modifiable contributor to patient-level heterogeneity in systemic inflammatory responses and clinical outcomes.


Subject(s)
Dysbiosis/etiology , Dysbiosis/mortality , Microbiota/genetics , Respiration, Artificial/adverse effects , Respiration, Artificial/mortality , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/mortality , Respiratory System/microbiology , Adult , Aged , Critical Illness/therapy , Female , Genetic Variation , Humans , Inflammation/etiology , Inflammation/microbiology , Male
3.
BMC Cancer ; 20(1): 383, 2020 May 06.
Article in English | MEDLINE | ID: mdl-32375706

ABSTRACT

BACKGROUND: The microbiome has been shown to affect the response to Immune Checkpoint Inhibitors (ICIs) in a small number of cancers and in preclinical models. Here, we sought to broadly survey cancers to identify those in which the microbiome may play a prognostic role using retrospective analyses of patients with advanced cancer treated with ICIs. METHODS: We conducted a retrospective analysis of 690 patients who received ICI therapy for advanced cancer. We used a literature review to define a causal model for the relationship between medications, the microbiome, and ICI response to guide the abstraction of electronic health records. Medications with precedent for changes to the microbiome included antibiotics, corticosteroids, proton pump inhibitors, histamine receptor blockers, non-steroid anti-inflammatories and statins. We tested the effect of medication timing on overall survival (OS) and evaluated the robustness of medication effects in each cancer. Finally, we compared the size of the effect observed for different classes of antibiotics to taxa that have been correlated to ICI response using a literature review of culture-based antibiotic susceptibilities. RESULTS: Of the medications assessed, only antibiotics and corticosteroids significantly associated with shorter OS. The hazard ratios (HRs) for antibiotics and corticosteroids were highest near the start of ICI treatment but remained significant when given prior to ICI. Antibiotics and corticosteroids remained significantly associated with OS even when controlling for multiple factors such as Eastern Cooperative Oncology Group performance status, Charlson Comorbidity Index score, and stage. When grouping antibiotics by class, ß-lactams showed the strongest association with OS across all tested cancers. CONCLUSIONS: The timing and strength of the correlations with antibiotics and corticosteroids after controlling for confounding factors are consistent with the microbiome involvement with the response to ICIs across several cancers.


Subject(s)
Adrenal Cortex Hormones/adverse effects , Anti-Bacterial Agents/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Bacteria/drug effects , Dysbiosis/mortality , Neoplasms/mortality , Dysbiosis/chemically induced , Dysbiosis/pathology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/pathology , Prognosis , Retrospective Studies , Survival Rate , Time Factors
4.
Int J Mol Med ; 45(4): 1130-1140, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32124946

ABSTRACT

The gut microbiota is important for maintaining the integrity of the intestinal barrier, promoting immunological tolerance and carrying out metabolic activities that have not evolved in hosts. Intestinal dysbiosis is associated with chronic kidney disease and probiotic supplementation has been shown to be beneficial. However, it is not known whether gut microorganisms­specifically, lactic acid bacteria (LAB) can protect against acute kidney injury (AKI). To address this issue, the present study investigated the effects of Lactobacillus salivarius BP121, an intestinal LAB isolated from the feces of newborns, in a rat model of cisplatin­induced AKI and also in Caco­2 human intestinal epithelial cells. BP121 prevented cisplatin­induced AKI in rats, as demonstrated by decreases in inflammation and oxidative stress in kidney tissue and in serum levels of uremic toxins such as indoxyl sulfate (IS) and p­cresol sulfate (PCS). BP121 also reduced intestinal permeability, as determined using fluorescein isothiocyanate­dextran by immunohistochemical detection of tight junction (TJ) proteins such as zona occludens­1 and occludin. The abundance of Lactobacillus spp., which are beneficial intestinal flora, was increased by BP121; this was accompanied by an increase in the concentrations of short­chain fatty acids in feces. Additionally, H2O2­induced TJ protein damage was reduced in Caco­2 cells treated with BP121 culture supernatant, an effect that was reversed by the 5' AMP­activated protein kinase (AMPK) inhibitor Compound C and Toll­like receptor (TLR)4 inhibitor TLR4­IN­C34. In conclusion, this study demonstrated that L. salivarius BP121 protects against cisplatin­induced AKI by decreasing inflammation and oxidative stress and this renoprotective effect is partially mediated by modulating the gut environment and thereby suppressing IS and PCS production as well as by regulating AMPK and TLR4 dependent TJ assembly.


Subject(s)
Acute Kidney Injury , Cisplatin/adverse effects , Cresols/metabolism , Dysbiosis , Indican/metabolism , Ligilactobacillus salivarius/metabolism , Sulfuric Acid Esters/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/microbiology , Acute Kidney Injury/prevention & control , Animals , Caco-2 Cells , Cisplatin/pharmacology , Dysbiosis/chemically induced , Dysbiosis/metabolism , Dysbiosis/mortality , Dysbiosis/prevention & control , Gastrointestinal Microbiome/drug effects , Humans , Male , Oxidative Stress , Rats , Rats, Sprague-Dawley
5.
Front Immunol ; 11: 589338, 2020.
Article in English | MEDLINE | ID: mdl-33574812

ABSTRACT

Beauveria bassiana is one of the most widespread insect pathogens and can be used in the biological control of agricultural, forestry and medical pests. The mechanisms by which B. bassiana leads to mortality in different host insects are also different. For house flies, B. bassiana has strong virulence, but its microecological mechanism is not clear. In this paper, the virulence of three strains of B. bassiana (TB, CB and BB) isolated from different hosts to house flies was studied. The results showed that the three strains of B. bassiana had strong pathogenicity to house fly adults. Specifically, TB was the strongest, CB was the second strongest, and BB was the weakest, with maximum lethal effects on house fly populations 5, 6, and 7 days after infection, respectively. Further study showed that the intestinal flora of house flies was disordered 3, 4, and 5 days after B. bassiana TB, CB and BB strain infection, respectively. Intestinal flora dysbacteriosis may be an important reason for the death of house flies caused by B. bassiana. After infection, the negative interaction ratio of bacteria in the house fly intestine decreased, and the stronger the virulence was, the lower the negative interaction ratio was. The time from B. bassiana infection to intestinal flora dysbacteriosis was not fixed. We named this period the "spring stage". The stronger the virulence of the B. bassiana strain was, the shorter the "spring stage" was. Therefore, the "spring stage" can be used as a virulence marker for evaluating the pathogenicity of different B. bassiana strains.


Subject(s)
Beauveria/pathogenicity , Dysbiosis/microbiology , Gastrointestinal Microbiome , Houseflies/microbiology , Mycoses/microbiology , Animals , Dysbiosis/mortality , Female , Gastrointestinal Microbiome/genetics , Male , Mycoses/mortality , Virulence
6.
Ann Oncol ; 30(10): 1572-1579, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31268133

ABSTRACT

Immune-checkpoint inhibitors (ICI) now represent the standard of care for several cancer types. In pre-clinical models, absence of an intact gut microbiome negatively impacted ICI efficacy and these findings permitted to unravel the importance of the commensal microbiota in immuno-oncology. Recently, multiple clinical studies including more than 1800 patients in aggregate demonstrated the negative predictive impact of treatments with broad-spectrum antibiotics (ATB) on cancer patients receiving ICI. Altogether, these results have led to the hypothesis that ATB-induced dysbiosis might influence the clinical response through the modulation of the gut microbiome. Controversy still remains, as ATB treatment might simply constitute a surrogate marker of unfit or immunodeficient patients. In this review, we summarize recent publications addressing the impact of the gut microbiome on ICI efficacy, discuss currently available data on the effect of ATB administered in different time-frames respect to ICI initiation, and finally, evoke the therapeutic implications of these findings.


Subject(s)
Anti-Bacterial Agents/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Dysbiosis/mortality , Gastrointestinal Microbiome/drug effects , Immunotherapy/mortality , Neoplasms/mortality , Dysbiosis/drug therapy , Dysbiosis/microbiology , Gastrointestinal Microbiome/immunology , Humans , Immunotherapy/adverse effects , Neoplasms/drug therapy , Neoplasms/pathology , Prognosis , Survival Rate
7.
Am J Respir Crit Care Med ; 199(10): 1205-1213, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30376356

ABSTRACT

Rationale: Lung dysbiosis promotes airway inflammation and decreased local immunity, potentially playing a role in the pathogenesis of acute exacerbations of chronic obstructive pulmonary disease (AECOPD). Objectives: We sought to determine the relationship between sputum microbiome at the time of AECOPD hospitalization and 1-year mortality in a COPD cohort. Methods: We used sputum samples from 102 patients hospitalized because of AECOPD. All subjects were followed for 1 year after discharge. The microbiome profile was assessed through sequencing of 16S rRNA gene. Microbiome analyses were performed according to 1-year mortality status. To investigate the effect of α-diversity measures and taxon features on time to death, we applied Cox proportional hazards regression models and obtained hazard ratios (HRs) associated with these variables. Measurements and Main Results: We observed significantly lower values of α-diversity (richness, Shannon index, evenness, and Faith's Phylogenetic Diversity) among nonsurvivors (n = 19, 18.6%) than survivors (n = 83, 81.4%). ß-Diversity analysis also demonstrated significant differences between both groups (adjusted permutational multivariate ANOVA, P = 0.010). The survivors had a higher relative abundance of Veillonella; in contrast, nonsurvivors had a higher abundance of Staphylococcus. The adjusted HRs for 1-year mortality increased significantly with decreasing α-diversity. We also observed lower survival among patients in whom sputum samples were negative for Veillonella (HR, 13.5; 95% confidence interval, 4.2-43.9; P < 0.001) or positive for Staphylococcus (HR, 7.3; 95% confidence interval, 1.6-33.2; P = 0.01). Conclusions: The microbiome profile of sputum in AECOPD is associated with 1-year mortality and may be used to identify subjects with a poor prognosis at the time of hospitalization.


Subject(s)
Dysbiosis/mortality , Microbiota , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/microbiology , Pulmonary Disease, Chronic Obstructive/mortality , Sputum/microbiology , Aged , British Columbia , Cohort Studies , Female , Hospitalization , Humans , Male , Middle Aged , Proportional Hazards Models
8.
Respir Res ; 19(1): 245, 2018 Dec 07.
Article in English | MEDLINE | ID: mdl-30526610

ABSTRACT

BACKGROUND: Host-associated microbial communities have important roles in tissue homeostasis and overall health. Severe perturbations can occur within these microbial communities during critical illness due to underlying diseases and clinical interventions, potentially influencing patient outcomes. We sought to profile the microbial composition of critically ill mechanically ventilated patients, and to determine whether microbial diversity is associated with illness severity and mortality. METHODS: We conducted a prospective, observational study of mechanically ventilated critically ill patients with a high incidence of pneumonia in 2 intensive care units (ICUs) in Hamilton, Canada, nested within a randomized trial for the prevention of healthcare-associated infections. The microbial profiles of specimens from 3 anatomical sites (respiratory, and upper and lower gastrointestinal tracts) were characterized using 16S ribosomal RNA gene sequencing. RESULTS: We collected 65 specimens from 34 ICU patients enrolled in the trial (29 endotracheal aspirates, 26 gastric aspirates and 10 stool specimens). Specimens were collected at a median time of 3 days (lower respiratory tract and gastric aspirates; interquartile range [IQR] 2-4) and 6 days (stool; IQR 4.25-6.75) following ICU admission. We observed a loss of biogeographical distinction between the lower respiratory tract and gastrointestinal tract microbiota during critical illness. Moreover, microbial diversity in the respiratory tract was inversely correlated with APACHE II score (r = - 0.46, p = 0.013) and was associated with hospital mortality (Median Shannon index: Discharged alive; 1.964 vs. Deceased; 1.348, p = 0.045). CONCLUSIONS: The composition of the host-associated microbial communities is severely perturbed during critical illness. Reduced microbial diversity reflects high illness severity and is associated with mortality. Microbial diversity may be a biomarker of prognostic value in mechanically ventilated patients. TRIAL REGISTRATION: ClinicalTrials.gov ID NCT01782755 . Registered February 4 2013.


Subject(s)
Dysbiosis/microbiology , Dysbiosis/mortality , Microbiological Phenomena , Respiration, Artificial/adverse effects , Respiration, Artificial/mortality , Aged , Critical Illness/epidemiology , Critical Illness/therapy , Dysbiosis/etiology , Female , Humans , Intensive Care Units/trends , Male , Middle Aged , Pilot Projects , Prospective Studies
9.
Article in English | MEDLINE | ID: mdl-30061286

ABSTRACT

Antibiotic disruption of the intestinal microbiota favors colonization by Clostridium difficile Using a charcoal-based adsorbent to decrease intestinal antibiotic concentrations, we studied the relationship between antibiotic concentrations in feces and the intensity of dysbiosis and quantified the link between this intensity and mortality. We administered either moxifloxacin (n = 70) or clindamycin (n = 60) to hamsters by subcutaneous injection from day 1 (D1) to D5 and challenged them with a C. difficile toxigenic strain at D3 Hamsters received various doses of a charcoal-based adsorbent, DAV131A, to modulate intestinal antibiotic concentrations. Gut dysbiosis was evaluated at D0 and D3 using diversity indices determined from 16S rRNA gene profiling. Survival was monitored until D16 We analyzed the relationship between fecal antibiotic concentrations and dysbiosis at the time of C. difficile challenge and studied their capacity to predict subsequent death of the animals. Increasing doses of DAV131A reduced fecal concentrations of both antibiotics, lowered dysbiosis, and increased survival from 0% to 100%. Mortality was related to the level of dysbiosis (P < 10-5 for the change of Shannon index in moxifloxacin-treated animals and P < 10-9 in clindamycin-treated animals). The Shannon diversity index and unweighted UniFrac distance best predicted death, with areas under the receiver operating curve (ROC) of 0.89 (95% confidence interval [CI], 0.82, 0.95) and 0.95 (0.90, 0.98), respectively. Altogether, moxifloxacin and clindamycin disrupted the diversity of the intestinal microbiota with a dependency on the DAV131A dose; mortality after C. difficile challenge was related to the intensity of dysbiosis in similar manners with the two antibiotics.


Subject(s)
Anti-Bacterial Agents/adverse effects , Clostridium Infections/drug therapy , Clostridium Infections/mortality , Dysbiosis/chemically induced , Animals , Anti-Bacterial Agents/therapeutic use , Clindamycin/therapeutic use , Clostridioides difficile/drug effects , Clostridioides difficile/pathogenicity , Cricetinae , Dysbiosis/mortality , Gastrointestinal Microbiome/drug effects , Male , Mesocricetus , Moxifloxacin/therapeutic use
10.
World J Gastroenterol ; 24(22): 2327-2347, 2018 Jun 14.
Article in English | MEDLINE | ID: mdl-29904241

ABSTRACT

Colorectal cancer (CRC) is the second leading cause of cancer worldwide. CRC is still associated with a poor prognosis among patients with advanced disease. On the contrary, due to its slow progression from detectable precancerous lesions, the prognosis for patients with early stages of CRC is encouraging. While most robust methods are invasive and costly, actual patient-friendly screening methods for CRC suffer of lack of sensitivity and specificity. Therefore, the development of sensitive, non-invasive and cost-effective methods for CRC detection and prognosis are necessary for increasing the chances of a cure. Beyond its beneficial functions for the host, increasing evidence suggests that the intestinal microbiota is a key factor associated with carcinogenesis. Many clinical studies have reported a disruption in the gut microbiota balance and an alteration in the faecal metabolome of CRC patients, suggesting the potential use of a microbial-based test as a non-invasive diagnostic and/or prognostic tool for CRC screening. This review aims to discuss the microbial signatures associated with CRC known to date, including dysbiosis and faecal metabolome alterations, and the potential use of microbial variation markers for non-invasive early diagnosis and/or prognostic assessment of CRC and advanced adenomas. We will finally discuss the possible use of these markers as predicators for treatment response and their limitations.


Subject(s)
Colorectal Neoplasms/diagnosis , Dysbiosis/microbiology , Early Detection of Cancer/methods , Gastrointestinal Microbiome/physiology , Biomarkers/analysis , Carcinogenesis/pathology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Dysbiosis/diagnosis , Dysbiosis/mortality , Dysbiosis/pathology , Feces/microbiology , Humans , Metabolome/physiology , Prognosis , Sensitivity and Specificity
11.
Ann Oncol ; 29(6): 1437-1444, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29617710

ABSTRACT

Background: The composition of gut microbiota affects antitumor immune responses, preclinical and clinical outcome following immune checkpoint inhibitors (ICI) in cancer. Antibiotics (ATB) alter gut microbiota diversity and composition leading to dysbiosis, which may affect effectiveness of ICI. Patients and methods: We examined patients with advanced renal cell carcinoma (RCC) and non-small-cell lung cancer (NSCLC) treated with anti-programmed cell death ligand-1 mAb monotherapy or combination at two academic institutions. Those receiving ATB within 30 days of beginning ICI were compared with those who did not. Objective response, progression-free survival (PFS) determined by RECIST1.1 and overall survival (OS) were assessed. Results: Sixteen of 121 (13%) RCC patients and 48 of 239 (20%) NSCLC patients received ATB. The most common ATB were ß-lactam or quinolones for pneumonia or urinary tract infections. In RCC patients, ATB compared with no ATB was associated with increased risk of primary progressive disease (PD) (75% versus 22%, P < 0.01), shorter PFS [median 1.9 versus 7.4 months, hazard ratio (HR) 3.1, 95% confidence interval (CI) 1.4-6.9, P < 0.01], and shorter OS (median 17.3 versus 30.6 months, HR 3.5, 95% CI 1.1-10.8, P = 0.03). In NSCLC patients, ATB was associated with similar rates of primary PD (52% versus 43%, P = 0.26) but decreased PFS (median 1.9 versus 3.8 months, HR 1.5, 95% CI 1.0-2.2, P = 0.03) and OS (median 7.9 versus 24.6 months, HR 4.4, 95% CI 2.6-7.7, P < 0.01). In multivariate analyses, the impact of ATB remained significant for PFS in RCC and for OS in NSCLC. Conclusion: ATB were associated with reduced clinical benefit from ICI in RCC and NSCLC. Modulatation of ATB-related dysbiosis and gut microbiota composition may be a strategy to improve clinical outcomes with ICI.


Subject(s)
Anti-Bacterial Agents/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Renal Cell/mortality , Dysbiosis/mortality , Kidney Neoplasms/mortality , Lung Neoplasms/mortality , Aged , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Cell Cycle Checkpoints/drug effects , Dysbiosis/chemically induced , Dysbiosis/pathology , Female , Follow-Up Studies , Humans , Immunotherapy/adverse effects , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Nivolumab/adverse effects , Prognosis , Survival Rate
12.
J Leukoc Biol ; 103(5): 799-805, 2018 05.
Article in English | MEDLINE | ID: mdl-29537705

ABSTRACT

Due to their cytotoxic activities, many anticancer drugs cause extensive damage to the intestinal mucosa and have antibiotic activities. Here, we show that cisplatin induces significant changes in the repertoire of intestinal commensal bacteria that exacerbate mucosal damage. Restoration of the microbiota through fecal-pellet gavage drives healing of cisplatin-induced intestinal damage. Bacterial translocation to the blood stream is correspondingly abrogated, resulting in a significant reduction in systemic inflammation, as evidenced by decreased serum IL-6 and reduced mobilization of granulocytes. Mechanistically, reversal of dysbiosis in response to fecal gavage results in the production of protective mucins and mobilization of CD11b+ myeloid cells to the intestinal mucosa, which promotes angiogenesis. Administration of Ruminococcus gnavus, a bacterial strain selectively depleted by cisplatin treatment, could only partially restore the integrity of the intestinal mucosa and reduce systemic inflammation, without measurable increases in the accumulation of mucin proteins. Together, our results indicate that reconstitution of the full repertoire of intestinal bacteria altered by cisplatin treatment accelerates healing of the intestinal epithelium and ameliorates systemic inflammation. Therefore, fecal microbiota transplant could paradoxically prevent life-threatening bacteremia in cancer patients treated with chemotherapy.


Subject(s)
Cisplatin/pharmacology , Dysbiosis/therapy , Fecal Microbiota Transplantation , Intestines/microbiology , Ovarian Neoplasms/drug therapy , Peritoneal Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Dysbiosis/mortality , Dysbiosis/pathology , Female , Gastrointestinal Microbiome , Intestines/drug effects , Intestines/pathology , Ovarian Neoplasms/microbiology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/microbiology , Peritoneal Neoplasms/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...