Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.050
Filter
1.
Microbiol Res ; 286: 127812, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38954992

ABSTRACT

Clostridioides difficile infection (CDI) poses a significant health threat due to high recurrence rates. Antimicrobial agents are commonly used to manage CDI-related diarrhoea; however, by aggravating intestinal dysbiosis, antibiotics enable C. difficile spores germination and production of toxins, the main virulence factors. Therefore, the binding of exotoxins using adsorbents represents an attractive alternative medication for the prevention and treatment of relapses. In this study, we provided evidence that the natural insoluble polysaccharides, named ABR119, extracted by plant cell cultures, effectively trap C. difficile toxins. In our experiments, ABR119 exhibited no cytotoxicity in vitro and was safely administered in vivo. In the animal model of C. difficile-associated colitis, ABR119 (50 mg/kg body weight) significantly reduced the colonic myeloperoxidase activity and severity of inflammation, preventing body weight loss. These effects were not evident when we treated animals with wheat bran polysaccharides. We did not detect bacterial killing effects of ABR119 against C. difficile nor against bacterial species of the normal gut microbiota. Moreover, ABR119 did not interfere in vitro with the antimicrobial activities of most clinically used antibiotics. In summary, ABR119 holds promise for treating and preventing C. difficile colitis by trapping the bacterial toxins, warranting further studies to assess the ABR119 potential in human infections caused by C. difficile.


Subject(s)
Anti-Bacterial Agents , Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Colitis , Disease Models, Animal , Polysaccharides , Polysaccharides/pharmacology , Polysaccharides/metabolism , Clostridioides difficile/drug effects , Clostridioides difficile/pathogenicity , Animals , Colitis/microbiology , Colitis/drug therapy , Colitis/prevention & control , Colitis/chemically induced , Clostridium Infections/prevention & control , Clostridium Infections/microbiology , Clostridium Infections/drug therapy , Bacterial Toxins/metabolism , Anti-Bacterial Agents/pharmacology , Humans , Plant Cells , Mice , Colon/microbiology , Colon/drug effects , Gastrointestinal Microbiome/drug effects
2.
Toxins (Basel) ; 16(6)2024 May 24.
Article in English | MEDLINE | ID: mdl-38922136

ABSTRACT

Clostridioides difficile, a Gram-positive anaerobic bacterium, is the leading cause of hospital-acquired antibiotic-associated diarrhea worldwide. The severity of C. difficile infection (CDI) varies, ranging from mild diarrhea to life-threatening conditions such as pseudomembranous colitis and toxic megacolon. Central to the pathogenesis of the infection are toxins produced by C. difficile, with toxin A (TcdA) and toxin B (TcdB) as the main virulence factors. Additionally, some strains produce a third toxin known as C. difficile transferase (CDT). Toxins damage the colonic epithelium, initiating a cascade of cellular events that lead to inflammation, fluid secretion, and further tissue damage within the colon. Mechanistically, the toxins bind to cell surface receptors, internalize, and then inactivate GTPase proteins, disrupting the organization of the cytoskeleton and affecting various Rho-dependent cellular processes. This results in a loss of epithelial barrier functions and the induction of cell death. The third toxin, CDT, however, functions as a binary actin-ADP-ribosylating toxin, causing actin depolymerization and inducing the formation of microtubule-based protrusions. In this review, we summarize our current understanding of the interaction between C. difficile toxins and host cells, elucidating the functional consequences of their actions. Furthermore, we will outline how this knowledge forms the basis for developing innovative, toxin-based strategies for treating and preventing CDI.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Host Microbial Interactions , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Clostridium Infections/pathology , Gene Order , Inflammation/pathology , Humans , Animals
3.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38928362

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a transcription factor that regulates the immune system through complicated transcriptional programs. Genistein, an AhR ligand, exhibits anti-inflammatory properties. However, its role in modulating immune responses via the AhR signaling pathway remains unclear. In this study, 360 male Arbor Acre broilers (1-day-old) were fed a basal diet supplemented with 40 or 80 mg/kg genistein and infected with or without Clostridium perfringens (Cp). Our results demonstrated that genistein ameliorated Cp-induced intestinal damage, as reflected by the reduced intestinal lesion scores and improved intestinal morphology and feed-to-gain ratio. Moreover, genistein increased intestinal sIgA, TGF-ß, and IL-10, along with elevated serum IgG, IgA, and lysozyme levels. Genistein improved intestinal AhR and cytochrome P450 family 1 subfamily A member 1 (CYP1A1) protein levels and AhR+ cell numbers in Cp-challenged broilers. The increased number of AhR+CD163+ cells in the jejunum suggested a potential association between genistein-induced AhR activation and anti-inflammatory effects mediated through M2 macrophage polarization. In IL-4-treated RAW264.7 cells, genistein increased the levels of AhR, CYP1A1, CD163, and arginase (Arg)-1 proteins, as well as IL-10 mRNA levels. This increase was attenuated by the AhR antagonist CH223191. In summary, genistein activated the AhR signaling pathway in M2 macrophages, which enhanced the secretion of anti-inflammatory cytokines and attenuated intestinal damage in Cp-infected broilers Cp.


Subject(s)
Chickens , Enteritis , Genistein , Macrophages , Receptors, Aryl Hydrocarbon , Animals , Receptors, Aryl Hydrocarbon/metabolism , Genistein/pharmacology , Genistein/therapeutic use , Macrophages/drug effects , Macrophages/metabolism , Mice , Enteritis/drug therapy , Enteritis/metabolism , Male , RAW 264.7 Cells , Poultry Diseases/drug therapy , Poultry Diseases/metabolism , Intestines/drug effects , Intestines/pathology , Clostridium perfringens , Clostridium Infections/drug therapy , Necrosis , Macrophage Activation/drug effects , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Signal Transduction/drug effects
4.
BMC Infect Dis ; 24(1): 625, 2024 Jun 23.
Article in English | MEDLINE | ID: mdl-38910242

ABSTRACT

BACKGROUND: In the last two decades, a significant increase in the number of Clostridioides difficile infection (CDI) cases has been observed. It is understandable to attempt to determine the factors that can predict the severity of the course of the infection and identify patients at risk of death. This study aimed to analyze the factors affecting the incidence and mortality of CDI in inpatient treatment at the University Clinical Hospital in Wroclaw in 2016-2018. METHODS: Statistical analysis of data obtained from patients' medical records was performed. Only patients with symptoms of infection and infection confirmed by laboratory tests were enrolled in the study. When analyzing the number of deaths, only adult patients who died in hospital wards were included. The quantitative data including laboratory tests, used antibiotics and Nutritional Risk Screening (NRS) were assessed. Also, the qualitative data such as sex, year of hospitalization, occurrence of diarrhoea on admission to the hospital, presence of additional diseases, as wee ad the use of antibacterial drugs or proton pump blockers and ranitidine during hospitalization were analyzed. RESULTS: A total of 319 adult CDI patients (178 women and 141 men) were enrolled of which 80 people died (50 women and 30 men). The mean age of the patients was 72.08 ± 16.74 years. Over the entire period studied, the morbidity was 174 cases per 100,000 hospitalizations while mortality was 25.08%. The group of deceased patients was characterized by: older age (by 9.24 years), longer duration of hospitalization (by 10 days), reduced albumin levels (Rho = -0.235, p < 0.001), higher urea levels, use of more antibiotics, higher risk of malnutrition in NRS (Rho = 0.219, p < 0.001), higher incidence of sepsis, heart failure, stroke, hypothyroidism. Pneumonia was diagnosed twice as often. It was also shown that deceased patients were significantly more likely to take penicillin and fluoroquinolones. CONCLUSIONS: In this study, the morbidity was lower, but mortality was higher compared to similar hospitals in Poland. CDI patients were characterized by older age, multimorbidity, extended hospitalization, and the use of broad-spectrum antibiotics. Risk factors for death included advanced age, prolonged hospital stays, lower albumin, higher urea, malnutrition, and comorbidities like heart failure, stroke, pneumonia, sepsis, and hypothyroidism. Increased antibiotic use, particularly penicillin and fluoroquinolones, was associated with a higher mortality risk.


Subject(s)
Clostridioides difficile , Clostridium Infections , Hospitalization , Hospitals, University , Humans , Male , Female , Clostridium Infections/mortality , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Clostridium Infections/drug therapy , Aged , Poland/epidemiology , Middle Aged , Aged, 80 and over , Hospitals, University/statistics & numerical data , Hospitalization/statistics & numerical data , Incidence , Risk Factors , Anti-Bacterial Agents/therapeutic use , Adult
5.
Nature ; 630(8016): 429-436, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38811738

ABSTRACT

Infections caused by Gram-negative pathogens are increasingly prevalent and are typically treated with broad-spectrum antibiotics, resulting in disruption of the gut microbiome and susceptibility to secondary infections1-3. There is a critical need for antibiotics that are selective both for Gram-negative bacteria over Gram-positive bacteria, as well as for pathogenic bacteria over commensal bacteria. Here we report the design and discovery of lolamicin, a Gram-negative-specific antibiotic targeting the lipoprotein transport system. Lolamicin has activity against a panel of more than 130 multidrug-resistant clinical isolates, shows efficacy in multiple mouse models of acute pneumonia and septicaemia infection, and spares the gut microbiome in mice, preventing secondary infection with Clostridioides difficile. The selective killing of pathogenic Gram-negative bacteria by lolamicin is a consequence of low sequence homology for the target in pathogenic bacteria versus commensals; this doubly selective strategy can be a blueprint for the development of other microbiome-sparing antibiotics.


Subject(s)
Anti-Bacterial Agents , Drug Discovery , Gastrointestinal Microbiome , Gram-Negative Bacteria , Gram-Negative Bacterial Infections , Symbiosis , Animals , Female , Humans , Male , Mice , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cell Line , Clostridioides difficile/drug effects , Clostridium Infections/microbiology , Clostridium Infections/drug therapy , Disease Models, Animal , Drug Design , Drug Resistance, Multiple, Bacterial , Gastrointestinal Microbiome/drug effects , Gram-Negative Bacteria/drug effects , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Lipoproteins/metabolism , Mice, Inbred C57BL , Protein Transport/drug effects , Sepsis/microbiology , Sepsis/drug therapy , Substrate Specificity , Symbiosis/drug effects
6.
Int J Antimicrob Agents ; 64(1): 107198, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734214

ABSTRACT

Clostridioides difficile (formerly Clostridium difficile) has been regarded as an 'urgent threat' and a significant global health problem, as life-threatening diarrhoea and refractory recurrence are common in patients with C. difficile infection (CDI). Unfortunately, the available anti-CDI drugs are limited. Recent guidelines recommend fidaxomicin and vancomycin as first-line drugs to treat CDI, bezlotoxumab to prevent recurrence, and faecal microbiota transplantation for rescue treatment. Currently, researchers are investigating therapeutic antibacterial drugs (e.g. teicoplanin, ridinilazole, ibezapolstat, surotomycin, cadazolid, and LFF571), preventive medications against recurrence (e.g. Rebyota, Vowst, VP20621, VE303, RBX7455, and MET-2), primary prevention strategies (e.g. vaccine, ribaxamase, and DAV132) and other anti-CDI medications in the preclinical stage (e.g. Raja 42, Myxopyronin B, and bacteriophage). This narrative review summarises current medications, including newly marketed drugs and products in development against CDI, to help clinicians treat CDI appropriately and to call for more research on innovation.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Fecal Microbiota Transplantation , Humans , Clostridium Infections/drug therapy , Clostridium Infections/prevention & control , Anti-Bacterial Agents/therapeutic use , Clostridioides difficile/drug effects , Vancomycin/therapeutic use , Fidaxomicin/therapeutic use
7.
N Z Med J ; 137(1594): 31-42, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38696830

ABSTRACT

AIM: Our antimicrobial guidelines (AGs) were changed in 2021 to recommend once-daily ceftriaxone in place of three-times-daily cefuroxime as preferred cephalosporin. This analysis sought to assess the effects of this on incidence of Clostridioides difficile infection (CDI), third-generation cephalosporin-resistant Enterobacterales (3GCR-E) and resource utilisation. METHOD: Before and after analysis of 30-day CDI and 3GCR-E incidence following receipt of cefuroxime/ceftriaxone pre- and post-AG change. Total nursing time and waste production relating to cefuroxime/ceftriaxone delivery were calculated pre- and post-change. RESULTS: CDI incidence was 0.6% pre- and 1.0% post-change (adjusted odds ratio [aOR] 1.44, p=0.07) and 3GCR-E incidence 3.5% and 3.1% (aOR 0.90, p=0.33). Mean per-quarter estimated nursing administration time decreased from 2,065 to 1,163 hours (902 nurse-hour reduction) and antibiotic-related waste generation from 1,131kg to 748kg (383kg reduction). Overall days of therapy per-quarter of cefuroxime/ceftriaxone were unchanged between periods. CONCLUSION: This simplification of our AG from a three-times-daily to a once-daily antibiotic resulted in considerable savings for our hospital (roughly 1.7 full-time equivalent nurses and over a tonne of waste yearly), with no significant increases in CDI or 3GCR-E. The impact of dosing schedules on non-antibiotic-spectrum factors, such as nursing time and resource usage, is worthy of consideration when designing AGs.


Subject(s)
Anti-Bacterial Agents , Antimicrobial Stewardship , Ceftriaxone , Cefuroxime , Humans , Cefuroxime/therapeutic use , Cefuroxime/administration & dosage , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/administration & dosage , Ceftriaxone/therapeutic use , Ceftriaxone/administration & dosage , Male , Female , Clostridium Infections/drug therapy , Clostridium Infections/epidemiology , Middle Aged , Incidence , Aged , Enterobacteriaceae Infections/drug therapy , Enterobacteriaceae Infections/epidemiology , Practice Guidelines as Topic , Drug Administration Schedule
8.
Gut Microbes ; 16(1): 2342583, 2024.
Article in English | MEDLINE | ID: mdl-38722061

ABSTRACT

Vancomycin and metronidazole are commonly used treatments for Clostridioides difficile infection (CDI). However, these antibiotics have been associated with high levels of relapse in patients. Fidaxomicin is a new treatment for CDI that is described as a narrow spectrum antibiotic that is minimally active on the commensal bacteria of the gut microbiome. The aim of this study was to compare the effect of fidaxomicin on the human gut microbiome with a number of narrow (thuricin CD) and broad spectrum (vancomycin and nisin) antimicrobials. The spectrum of activity of each antimicrobial was tested against 47 bacterial strains by well-diffusion assay. Minimum inhibitory concentrations (MICs) were calculated against a select number of these strains. Further, a pooled fecal slurry of 6 donors was prepared and incubated for 24 h with 100 µM of each antimicrobial in a mini-fermentation system together with a no-treatment control. Fidaxomicin, vancomycin, and nisin were active against most gram positive bacteria tested in vitro, although fidaxomicin and vancomycin produced larger zones of inhibition compared to nisin. In contrast, the antimicrobial activity of thuricin CD was specific to C. difficile and some Bacillus spp. The MICs showed similar results. Thuricin CD exhibited low MICs (<3.1 µg/mL) for C. difficile and Bacillus firmus, whereas fidaxomicin, vancomycin, and nisin demonstrated lower MICs for all other strains tested when compared to thuricin CD. The narrow spectrum of thuricin CD was also observed in the gut model system. We conclude that the spectrum of activity of fidaxomicin is comparable to that of the broad-spectrum antibiotic vancomycin in vitro and the broad spectrum bacteriocin nisin in a complex community.


Subject(s)
Anti-Bacterial Agents , Feces , Fidaxomicin , Gastrointestinal Microbiome , Microbial Sensitivity Tests , Nisin , Vancomycin , Nisin/pharmacology , Anti-Bacterial Agents/pharmacology , Humans , Fidaxomicin/pharmacology , Vancomycin/pharmacology , Gastrointestinal Microbiome/drug effects , Feces/microbiology , Bacteria/drug effects , Bacteria/classification , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Bacteriocins/pharmacology
9.
J Med Chem ; 67(11): 9465-9484, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38753983

ABSTRACT

Clostridioides difficile (C. difficile) is one of the leading causes of healthcare-associated infections worldwide. The increasing incidence of strains resistant to currently available therapies highlights the need for alternative treatment options with a novel mode of action. Oxazolidinones that are connected to a quinolone moiety with a pyrrolidine linker, such as compound 1, are reported to exhibit potent broadspectrum antibacterial activity. In an effort to optimize this class of compounds for the treatment of C. difficile infection (CDI), we have identified cadazolid (9), a first-in-class quinoxolidinone antibiotic, which is a potent inhibitor of C. difficile protein synthesis. In order to achieve narrow-spectrum coverage of clinically most relevant strains without affecting the gut microbiota, an emphasis was placed on abolishing activity against commensals of the intestinal microbiome while retaining good coverage of pathogenic C. difficile, including hypervirulent and epidemic strains.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Microbial Sensitivity Tests , Structure-Activity Relationship , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/chemical synthesis , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Animals , Humans , Drug Discovery , Gastrointestinal Microbiome/drug effects , Mice , Oxazolidinones
10.
J Microbiol Biotechnol ; 34(4): 828-837, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38668685

ABSTRACT

Vancomycin (VAN) and metronidazole (MTR) remain the current drugs of choice for the treatment of non-severe Clostridioides difficile infection (CDI); however, while their co-administration has appeared in clinical treatment, the efficacy varies greatly and the mechanism is unknown. In this study, a CDI mouse model was constructed to evaluate the therapeutic effects of VAN and MTR alone or in combination. For a perspective on the intestinal ecology, 16S rRNA amplicon sequencing and non-targeted metabolomics techniques were used to investigate changes in the fecal microbiota and metabolome of mice under the co-administration treatment. As a result, the survival rate of mice under co-administration was not dramatically different compared to that of single antibiotics, and the former caused intestinal tissue hyperplasia and edema. Co-administration also significantly enhanced the activity of amino acid metabolic pathways represented by phenylalanine, arginine, proline, and histidine, decreased the level of deoxycholic acid (DCA), and downregulated the abundance of beneficial microbes, such as Bifidobacterium and Akkermansia. VAN plays a dominant role in microbiota regulation in co-administration. In addition, co-administration reduced or increased the relative abundance of antibiotic-sensitive bacteria, including beneficial and harmful microbes, without a difference. Taken together, there are some risks associated with the co-administration of VAN and MTR, and this combination mode should be used with caution in CDI treatment.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Disease Models, Animal , Drug Therapy, Combination , Feces , Gastrointestinal Microbiome , Metronidazole , RNA, Ribosomal, 16S , Vancomycin , Animals , Metronidazole/administration & dosage , Vancomycin/administration & dosage , Vancomycin/pharmacology , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Gastrointestinal Microbiome/drug effects , Mice , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , RNA, Ribosomal, 16S/genetics , Feces/microbiology , Intestines/microbiology , Intestines/drug effects , Male , Bacteria/classification , Bacteria/genetics , Bacteria/drug effects , Metabolome/drug effects
11.
J Antimicrob Chemother ; 79(6): 1413-1417, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38661207

ABSTRACT

OBJECTIVES: To assess the effectiveness of shortened regimens of vancomycin or fidaxomicin in the treatment of Clostridioides difficile infection (CDI). METHODS: Adult patients with CDI hospitalized from January 2022 to May 2023 were included in this observational study. In patients with CDI treated with vancomycin or fidaxomicin, antibiotic treatment was discontinued after either 5 or 7 days of vancomycin or 5 days of fidaxomicin if there was a clinical response and improvement in laboratory parameters. The control cohort was treated with the standard 10 day regimen of either vancomycin or fidaxomicin. The follow-up was 60 days. Causative C. difficile strains were characterized by ribotyping and toxin gene detection when available. RESULTS: Twenty-five patients (median age 76 years) received shortened treatment with vancomycin (n = 21), or fidaxomicin (n = 4). Five cases fulfilled the criteria for severe CDI. Twenty-three patients completed follow-up; two died from causes other than CDI, and two developed recurrent CDI (8.0%). Ribotypes (RTs) 001 and 014 were the most prevalent with 20% each. In two C. difficile isolates, binary toxin genes were detected (RTs 078 and 023). In the control group of 22 patients recurrent CDI developed in 5 patients (22.7%). No statistically significant differences were found between the groups. CONCLUSIONS: Shortened treatment regimens for CDI with vancomycin and fidaxomicin were shown to be effective in our cohort of patients compared with 10 days of treatment. The recurrence rate was lower in the study group. A larger, prospective, double-blind, randomized, multicentre study is needed to support our findings.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Fidaxomicin , Ribotyping , Vancomycin , Humans , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Aged , Male , Female , Clostridioides difficile/genetics , Clostridioides difficile/drug effects , Clostridioides difficile/classification , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/administration & dosage , Vancomycin/therapeutic use , Vancomycin/administration & dosage , Fidaxomicin/therapeutic use , Fidaxomicin/administration & dosage , Middle Aged , Aged, 80 and over , Treatment Outcome
12.
EBioMedicine ; 103: 105130, 2024 May.
Article in English | MEDLINE | ID: mdl-38653188

ABSTRACT

BACKGROUND: Active surveillance pharmacovigilance is an emerging approach to identify medications with unanticipated effects. We previously developed a framework called pharmacopeia-wide association studies (PharmWAS) that limits false positive medication associations through high-dimensional confounding adjustment and set enrichment. We aimed to assess the transportability and generalizability of the PharmWAS framework by using medical claims data to reproduce known medication associations with Clostridioides difficile infection (CDI) or gastrointestinal bleeding (GIB). METHODS: We conducted case-control studies using Optum's de-identified Clinformatics Data Mart Database of individuals enrolled in large commercial and Medicare Advantage health plans in the United States. Individuals with CDI (from 2010 to 2015) or GIB (from 2010 to 2021) were matched to controls by age and sex. We identified all medications utilized prior to diagnosis and analysed the association of each with CDI or GIB using conditional logistic regression adjusted for risk factors for the outcome and a high-dimensional propensity score. FINDINGS: For the CDI study, we identified 55,137 cases, 220,543 controls, and 290 medications to analyse. Antibiotics with Gram-negative spectrum, including ciprofloxacin (aOR 2.83), ceftriaxone (aOR 2.65), and levofloxacin (aOR 1.60), were strongly associated. For the GIB study, we identified 450,315 cases, 1,801,260 controls, and 354 medications to analyse. Antiplatelets, anticoagulants, and non-steroidal anti-inflammatory drugs, including ticagrelor (aOR 2.81), naproxen (aOR 1.87), and rivaroxaban (aOR 1.31), were strongly associated. INTERPRETATION: These studies demonstrate the generalizability and transportability of the PharmWAS pharmacovigilance framework. With additional validation, PharmWAS could complement traditional passive surveillance systems to identify medications that unexpectedly provoke or prevent high-impact conditions. FUNDING: U.S. National Institute of Diabetes and Digestive and Kidney Diseases.


Subject(s)
Clostridioides difficile , Clostridium Infections , Gastrointestinal Hemorrhage , Pharmacovigilance , Humans , Clostridium Infections/epidemiology , Clostridium Infections/etiology , Clostridium Infections/drug therapy , Case-Control Studies , Male , Gastrointestinal Hemorrhage/chemically induced , Gastrointestinal Hemorrhage/epidemiology , Gastrointestinal Hemorrhage/etiology , Female , Aged , Middle Aged , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , United States/epidemiology , Risk Factors , Adult , Aged, 80 and over
13.
J Antimicrob Chemother ; 79(6): 1320-1328, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38598696

ABSTRACT

OBJECTIVES: To determine the frequencies and clonal distributions of putative genetic determinants of resistance to antimicrobials applied for treatment of Clostridioides difficile infection (CDI), as documented in the genomic record. METHODS: We scanned 26 557 C. difficile genome sequences publicly available from the EnteroBase platform for plasmids, point mutations and gene truncations previously reported to reduce susceptibility to vancomycin, fidaxomicin or metronidazole, respectively. We measured the antimicrobial susceptibility of 143 selected C. difficile isolates. RESULTS: The frequency of mutations causing reduced susceptibility to vancomycin and metronidazole, respectively, increased strongly after 2000, peaking at up to 52% of all sequenced C. difficile genomes. However, both mutations declined sharply more recently, reflecting major changes in CDI epidemiology. We detected mutations associated with fidaxomicin resistance in several major genotypes, but found no evidence of international spread of resistant clones. The pCD-METRO plasmid, conferring metronidazole resistance, was detected in a single previously unreported C. difficile isolate, recovered from a hospital patient in Germany in 2008. The pX18-498 plasmid, putatively associated with decreased vancomycin susceptibility, was confined to related, recent isolates from the USA. Phenotype measurements confirmed that most of those genetic features were useful predictors of antibiotic susceptibility, even though ranges of MICs typically overlapped among isolates with and without specific mutations. CONCLUSIONS: Genomic data suggested that resistance to therapeutic antimicrobial drugs is rare in C. difficile. Public antimicrobial resistance marker databases were not equipped to detect most of the genetic determinants relevant to antibiotic therapy of CDI.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Drug Resistance, Bacterial , Genome, Bacterial , Microbial Sensitivity Tests , Plasmids , Clostridioides difficile/genetics , Clostridioides difficile/drug effects , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Bacterial/genetics , Plasmids/genetics , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Vancomycin/pharmacology , Vancomycin/therapeutic use , Metronidazole/pharmacology , Metronidazole/therapeutic use , Genotype , Mutation
14.
Ann Clin Microbiol Antimicrob ; 23(1): 35, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664689

ABSTRACT

PURPOSE: The clinical significance of negative toxin enzyme immunoassays (EIA) for Clostridioides difficile infections (CDIs) is unclear. Our study aimed to investigate the significance of toxin EIA-negative in the diagnosis and prognosis of CDI. METHODS: All stool specimens submitted for C. difficile toxin EIA testing were cultured to isolate C. difficile. In-house PCR for tcdA, tcdB, cdtA, and cdtB genes were performed using C. difficile isolates. Stool specimens were tested with C. difficile toxins A and B using EIA kit (RIDASCREEN Clostridium difficile toxin A/B, R-Biopharm AG, Darmstadt, Germany). Characteristics and subsequent CDI episodes of toxin EIA-negative and -positive patients were compared. RESULTS: Among 190 C. difficile PCR-positive patients, 83 (43.7%) were toxin EIA-negative. Multivariate analysis revealed independent associations toxin EIA-negative results and shorter hospital stays (OR = 0.98, 95% CI 0.96-0.99, p = 0.013) and less high-risk antibiotic exposure in the preceding month (OR = 0.38, 95% CI 0.16-0.94, p = 0.035). Toxin EIA-negative patients displayed a significantly lower white blood cell count rate (11.0 vs. 35.4%, p < 0.001). Among the 54 patients who were toxin EIA-negative and did not receive CDI treatment, three (5.6%) were diagnosed with CDI after 7-21 days without complication. CONCLUSION: Our study demonstrates that toxin EIA-negative patients had milder laboratory findings and no complications, despite not receiving treatment. Prolonged hospitalisation and exposure to high-risk antibiotics could potentially serve as markers for the development of toxin EIA-positive CDI.


Subject(s)
Bacterial Proteins , Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Feces , Humans , Clostridioides difficile/genetics , Feces/microbiology , Male , Female , Bacterial Toxins/analysis , Clostridium Infections/diagnosis , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Aged , Middle Aged , Bacterial Proteins/genetics , Bacterial Proteins/analysis , Enterotoxins/analysis , Aged, 80 and over , Anti-Bacterial Agents/therapeutic use , Immunoenzyme Techniques , Adult , Treatment Outcome , Polymerase Chain Reaction , Prognosis
15.
Fitoterapia ; 176: 105973, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38663560

ABSTRACT

The growing incidence of Clostridium difficile associated diarrhea (CDAD) underscores the urgency for potent treatments. This research delves into the therapeutic potential of Scutellaria baicalensis Georgi (Lamiaceae) root (SR) in addressing CDAD and its influence on gut microbiota. Using a CDAD mouse model and fidaxomicin as a control, SR's impact was measured through diarrhea symptoms, colonic histopathology, and C. difficile toxin levels. Employing the PacBio platform, 16S rRNA full-length gene sequencing analyzed the gut microbial composition and the effect of SR. Results revealed SR considerably alleviated diarrhea during treatment and restoration phases, with a marked decrease in colonic inflammation. C. difficile toxin levels dropped significantly with SR treatment (P < 0.001). While SR didn't augment gut microbiota's overall abundance, it enhanced its diversity. It restored levels of Proteobacteria and Bacteroidetes, reduced Akkermansia spp. and Enterococcus spp. proportions, and modulated specific bacterial species' abundance. In essence, SR effectively mitigates CDAD symptoms, curtails inflammatory reactions, and beneficially restructures gut microbiota, suggesting its potential in advanced CDAD clinical intervention.


Subject(s)
Clostridioides difficile , Diarrhea , Gastrointestinal Microbiome , Plant Extracts , Scutellaria baicalensis , Gastrointestinal Microbiome/drug effects , Animals , Diarrhea/microbiology , Diarrhea/drug therapy , Mice , Scutellaria baicalensis/chemistry , Plant Extracts/pharmacology , Plant Roots/chemistry , Male , Clostridium Infections/drug therapy , Disease Models, Animal , RNA, Ribosomal, 16S/genetics , Mice, Inbred C57BL , Colon/microbiology
16.
Rinsho Ketsueki ; 65(3): 153-157, 2024.
Article in Japanese | MEDLINE | ID: mdl-38569858

ABSTRACT

An 80-year-old man with FLT3-TKD mutation-positive acute myeloid leukemia (AML) relapsed during consolidation therapy with venetoclax/azacitidine and was started on gilteritinib as salvage therapy. On the day after treatment initiation, febrile neutropenia was observed, but the fever resolved promptly after initiation of antimicrobial therapy. On the fifth day after completion of antimicrobial therapy, the patient experienced fever and watery diarrhea over 10 times a day, and a diagnosis of Clostridioides difficile infection (CDI) was made based on stool examination. The patient was treated with intravenous metronidazole, but renal dysfunction, hypotension, and hypoxemia developed, and a CT scan showed pleural and intraperitoneal effusion, significant intestinal wall thickening, and intestinal dilatation. Fidaxomicin was started under general monitoring in the intensive care unit and response was achieved. The patient was discharged from the intensive care unit on the 18th day after the onset of CDI. We report this case not only due to the rarity of fulminant CDI during AML treatment, but also because it is a valuable example of effective treatment of fulminant CDI with fidaxomicin.


Subject(s)
Anti-Infective Agents , Clostridium Infections , Leukemia, Myeloid, Acute , Male , Humans , Aged, 80 and over , Fidaxomicin , Clostridium Infections/drug therapy , Treatment Outcome , Protein Kinase Inhibitors , Leukemia, Myeloid, Acute/drug therapy , Anti-Bacterial Agents/adverse effects , fms-Like Tyrosine Kinase 3
17.
Rev. chil. infectol ; 41(2): 193-198, abr. 2024. ilus, tab
Article in Spanish | LILACS | ID: biblio-1559684

ABSTRACT

INTRODUCCIÓN: En la diarrea asociada a Clostridioides dfficile (DACD) leve-moderada se recomienda tratar con vancomicina por sobre metronidazol, a pesar de su difícil acceso y poca evidencia en el medio ambulatorio. OBJETIVO: Comparar la tasa de cura clínica y recurrencia entre vancomicina y metronidazol en adultos chilenos con primer episodio leve-moderado de DACD de manejo ambulatorio. MÉTODOS: Cohorte retrospectiva entre enero 2015 y diciembre 2020 en centros de una red de salud universitaria de pacientes de ≥ 18 años con DACD tratados ambulatoriamente. RESULTADOS: Se obtuvieron 161 pacientes, 59% mujeres, edad promedio de 53 años (entre 18 y 94 años). De ellos, 109 (67,7%) usaron metronidazol y 52 (32,3%) vancomicina. En el análisis multivariado ajustado por edad y comorbilidades se obtuvo un OR 3,00 (IC 95% 1,12-9,59) para cura clínica y 0,27 (IC 95% 0,06-0,88) para recurrencia a ocho semanas, ambos a favor de vancomicina, sin diferencias en recurrencia a 12 meses, necesidad de hospitalización o mortalidad. CONCLUSIÓN: La terapia con vancomicina comparada contra metronidazol en el tratamiento ambulatorio de la infección leve-moderada por C. dfficile se asocia a mayor cura clínica y menor tasa de recurrencia a corto plazo, sin diferencias en desenlaces a largo plazo.


BACKGROUND: Recommended treatment against mild cases of Clostridioides difficile associated diarrhea is vancomycin despite the difficulties of access compared to metronidazole. AIM: To compare the effectiveness of vancomycin and metronidazole in Chilean adults with first mild-moderate episode of Clostridiodes difficile infection (CDI). METHODS: Retrospective cohort of patients with CDI between January 2015 and December 2020 treated in centers of a university health network. The patients were adults treated for C. difficile infection on an outpatient basis. Recurrent and severe cases were excluded. Outcomes included clinical cure and recurrence rate. RESULTS: Data from 161 patients was recovered. Fifty-nine percent were women and average age was 53 (18-94). One hundred and nine patients were treated with metronidazole (67.7%) and 52 (32.3%) used vancomycin. Multivariate analysis adjusted by age and comorbidities showed an Odds Ratio of 3.00 (IC 95% 1.12-9.59) for clinical cure and 0.27 (IC 95% 0.06-0.88) for 8-week recurrence rate, both in favor of vancomycin, without differences in 12-month recurrence rate, hospitalization rate nor mortality. CONCLUSIONS: Vancomycin is associated with better short-term outcomes in the treatment of outpatient mild-moderate first episode C. difficile infection, without differences in long term recurrence or mortality when compared with metronidazole.


Subject(s)
Humans , Male , Female , Adolescent , Adult , Middle Aged , Aged , Aged, 80 and over , Young Adult , Vancomycin/therapeutic use , Clostridium Infections/drug therapy , Diarrhea/drug therapy , Metronidazole/therapeutic use , Outpatients , Recurrence , Multivariate Analysis , Regression Analysis , Retrospective Studies , Ambulatory Care , Anti-Bacterial Agents/therapeutic use
18.
Gut Microbes ; 16(1): 2337312, 2024.
Article in English | MEDLINE | ID: mdl-38591915

ABSTRACT

Clostridioides difficile causes a range of debilitating intestinal symptoms that may be fatal. It is particularly problematic as a hospital-acquired infection, causing significant costs to the health care system. Antibiotics, such as vancomycin and fidaxomicin, are still the drugs of choice for C. difficile infections, but their effectiveness is limited, and microbial interventions are emerging as a new treatment option. This paper focuses on alternative treatment approaches, which are currently in various stages of development and can be divided into four therapeutic strategies. Direct killing of C. difficile (i) includes beside established antibiotics, less studied bacteriophages, and their derivatives, such as endolysins and tailocins. Restoration of microbiota composition and function (ii) is achieved with fecal microbiota transplantation, which has recently been approved, with standardized defined microbial mixtures, and with probiotics, which have been administered with moderate success. Prevention of deleterious effects of antibiotics on microbiota is achieved with agents for the neutralization of antibiotics that act in the gut and are nearing regulatory approval. Neutralization of C. difficile toxins (iii) which are crucial virulence factors is achieved with antibodies/antibody fragments or alternative binding proteins. Of these, the monoclonal antibody bezlotoxumab is already in clinical use. Immunomodulation (iv) can help eliminate or prevent C. difficile infection by interfering with cytokine signaling. Small-molecule agents without bacteriolytic activity are usually selected by drug repurposing and can act via a variety of mechanisms. The multiple treatment options described in this article provide optimism for the future treatment of C. difficile infection.


Subject(s)
Clostridioides difficile , Clostridium Infections , Gastrointestinal Microbiome , Humans , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Fecal Microbiota Transplantation , Vancomycin/pharmacology , Clostridium Infections/drug therapy , Clostridium Infections/prevention & control
20.
Am J Med ; 137(7): 571-576, 2024 07.
Article in English | MEDLINE | ID: mdl-38508330

ABSTRACT

Clostridioides difficile infection is the most common healthcare-associated infection in the United States, with potential life-threatening complications and a significant impact on the costs of care. Antibiotic stewardship as well as discontinuation of chronic acid suppressive therapy are key for its prevention and treatment. Effective infection management requires appropriate interpretation of diagnostic tests, as well as the use of vancomycin and fidaxomicin as first-line treatment. Novel treatments such as Bezlotoxumab, fecal microbiota transplant, and live biotherapeutic products are proven effective in recurrent C. difficile infection and address dysbiosis.


Subject(s)
Anti-Bacterial Agents , Clostridium Infections , Fecal Microbiota Transplantation , Humans , Clostridium Infections/diagnosis , Clostridium Infections/therapy , Clostridium Infections/drug therapy , Anti-Bacterial Agents/therapeutic use , Clostridioides difficile , Antimicrobial Stewardship , Vancomycin/therapeutic use , Fidaxomicin/therapeutic use , Broadly Neutralizing Antibodies , Antibodies, Monoclonal
SELECTION OF CITATIONS
SEARCH DETAIL