Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.782
Filter
1.
Nat Chem Biol ; 20(7): 924-933, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38942968

ABSTRACT

Keratinicyclins and keratinimicins are recently discovered glycopeptide antibiotics. Keratinimicins show broad-spectrum activity against Gram-positive bacteria, while keratinicyclins form a new chemotype by virtue of an unusual oxazolidinone moiety and exhibit specific antibiosis against Clostridioides difficile. Here we report the mechanism of action of keratinicyclin B (KCB). We find that steric constraints preclude KCB from binding peptidoglycan termini. Instead, KCB inhibits C. difficile growth by binding wall teichoic acids (WTAs) and interfering with cell wall remodeling. A computational model, guided by biochemical studies, provides an image of the interaction of KCB with C. difficile WTAs and shows that the same H-bonding framework used by glycopeptide antibiotics to bind peptidoglycan termini is used by KCB for interacting with WTAs. Analysis of KCB in combination with vancomycin (VAN) shows highly synergistic and specific antimicrobial activity, and that nanomolar combinations of the two drugs are sufficient for complete growth inhibition of C. difficile, while leaving common commensal strains unaffected.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Microbial Sensitivity Tests , Clostridioides difficile/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Vancomycin/pharmacology , Vancomycin/chemistry , Cell Wall/drug effects , Cell Wall/metabolism , Teichoic Acids/metabolism , Peptidoglycan/metabolism , Peptidoglycan/chemistry , Drug Therapy, Combination , Peptides, Cyclic , Lipopeptides
2.
Braz J Infect Dis ; 28(3): 103767, 2024.
Article in English | MEDLINE | ID: mdl-38843868

ABSTRACT

BACKGROUND: C. difficile has been increasingly reported as a cause of gastrointestinal disease in children, ranging from mild self-limiting diarrhea to severe conditions such as pseudomembranous colitis and toxic megacolon. Only two pediatric research groups reported the presence of C. difficile infection in Brazilian children, but no previous research has examined C. difficile infection among children in northeastern Brazil. This prospective cross-sectional study investigated the molecular epidemiology and antimicrobial resistance of C. difficile strains isolated from children and adolescents with diarrhea referred to a tertiary pediatric hospital in Brazil while exploring the associated risk factors. RESULTS: Toxin positivity or C. difficile isolation was found in 30.4 % (17/56) samples. C. difficile was isolated from 35 % (6/17) samples. Four toxigenic strains were identified (tpi+, tcdA+, tcdB+, cdtB-, without tcdC deletions) belonging to PCR ribotypes and PFGE-pulsotypes: 046 (new pulsotype 1174), 106 (NAP11), 002 (new pulsotype 1274), 012 (new pulsotype NML-1235). Two of the six isolates belonging to ribotypes 143 and 133 were non-toxigenic. All toxigenic strains were sensitive to metronidazole and vancomycin. Regarding the clinical manifestation, diarrhea lasted an average of 11 days, ranging from 3 to 50 days and was often associated with mucus and/or blood. All six patients from whom the C. difficile was isolated had a chronic disease diagnosis, with these comorbidities as the main risk factors. CONCLUSION: Our study enhances our understanding of the present epidemiological landscape of C. difficile-associated diarrhea (CDI) among children in northeastern Brazil, reveling a substantial CDI frequency of 30.4 %, with toxigenic strains detected in 76.4 % of cases, highlighting a higher prevalence compared to earlier Brazilian studies. In the globalized world, an understanding of disease-generating strains, the associated risk factors, clinical manifestation, and antimicrobial sensitivity has fundamental epidemiological importance and draws attention to preventive measures, allowing for more decisive action.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Hospitals, Pediatric , Microbial Sensitivity Tests , Tertiary Care Centers , Humans , Clostridioides difficile/genetics , Clostridioides difficile/drug effects , Clostridioides difficile/isolation & purification , Child , Adolescent , Female , Male , Brazil/epidemiology , Cross-Sectional Studies , Prospective Studies , Tertiary Care Centers/statistics & numerical data , Child, Preschool , Anti-Bacterial Agents/pharmacology , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Risk Factors , Infant , Molecular Epidemiology , Diarrhea/microbiology , Diarrhea/epidemiology , Ribotyping , Drug Resistance, Bacterial/genetics
3.
J Hosp Infect ; 149: 22-25, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38705474

ABSTRACT

BACKGROUND: The role of the healthcare environment in the transmission of clinical pathogens is well established. EN 17126:2018 was developed to address the need for regulated sporicidal product testing and includes a realistic medical soil to enable validation of products that claim combined cleaning and disinfection efficacy. AIM: To investigate the chemical stability and sporicidal efficacy of oxidizing disinfectant products in the presence of simulated clean and medical dirty conditions. METHODS: Disinfectant stability and sporicidal efficacy were evaluated in like-for-like ratios of soil:product. Disinfectants were exposed to simulated test soils and free chlorine, chlorine dioxide or peracetic acid concentrations were measured using standard colorimetric methods. Efficacy of disinfectants against C. difficile R027 endospores was assessed as per EN 17126:2018. Comparisons of performance between clean and medical dirty conditions were performed using one-way analysis of variance. Correlation analysis was performed using Pearson product-moment correlation. FINDINGS: Performance of chlorine-releasing agents (sodium dichloroisocyanurate, chlorine dioxide and hypochlorous acid) was concentration dependent, with 1000 ppm chlorine showing reduced stability and efficacy in dirty conditions. By contrast, peracetic acid product demonstrated stability and consistently achieved efficacy in dirty conditions. CONCLUSION: These results have implications for clinical practice, as ineffective environmental decontamination may increase the risk of transmission of pathogens that can cause healthcare-associated infections.


Subject(s)
Chlorine Compounds , Disinfectants , Oxides , Peracetic Acid , Spores, Bacterial , Disinfectants/pharmacology , Chlorine Compounds/pharmacology , Oxides/pharmacology , Peracetic Acid/pharmacology , Spores, Bacterial/drug effects , Clostridioides difficile/drug effects , Humans , Disinfection/methods , Triazines/pharmacology , Hypochlorous Acid/pharmacology
4.
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
5.
Microb Genom ; 10(5)2024 May.
Article in English | MEDLINE | ID: mdl-38717815

ABSTRACT

Clostridioides difficile infection (CDI) remains a significant public health threat globally. New interventions to treat CDI rely on an understanding of the evolution and epidemiology of circulating strains. Here we provide longitudinal genomic data on strain diversity, transmission dynamics and antimicrobial resistance (AMR) of C. difficile ribotypes (RTs) 014/020 (n=169), 002 (n=77) and 056 (n=36), the three most prominent C. difficile strains causing CDI in Australia. Genome scrutiny showed that AMR was uncommon in these lineages, with resistance-conferring alleles present in only 15/169 RT014/020 strains (8.9 %), 1/36 RT056 strains (2.78 %) and none of 77 RT002 strains. Notably, ~90 % of strains were resistant to MLSB agents in vitro, but only ~5.9 % harboured known resistance alleles, highlighting an incongruence between AMR genotype and phenotype. Core genome analyses revealed all three RTs contained genetically heterogeneous strain populations with limited evidence of clonal transmission between CDI cases. The average number of pairwise core genome SNP (cgSNP) differences within each RT group ranged from 23.3 (RT056, ST34, n=36) to 115.6 (RT002, ST8, n=77) and 315.9 (RT014/020, STs 2, 13, 14, 49, n=169). Just 19 clonal groups (encompassing 40 isolates), defined as isolates differing by ≤2 cgSNPs, were identified across all three RTs (RT014/020, n=14; RT002, n=3; RT056, n=2). Of these clonal groups, 63 % (12/19) comprised isolates from the same Australian State and 37 % (7/19) comprised isolates from different States. The low number of plausible transmission events found for these major RTs (and previously documented populations in animal and environmental sources/reservoirs) points to widespread and persistent community sources of diverse C. difficile strains as opposed to ongoing nationwide healthcare outbreaks dominated by a single clone. Together, these data provide new insights into the evolution of major lineages causing CDI in Australia and highlight the urgent need for enhanced surveillance, and for public health interventions to move beyond the healthcare setting and into a One Health paradigm to effectively combat this complex pathogen.


Subject(s)
Clostridioides difficile , Clostridium Infections , Phylogeny , Ribotyping , Clostridioides difficile/genetics , Clostridioides difficile/classification , Clostridioides difficile/drug effects , Clostridioides difficile/isolation & purification , Australia/epidemiology , Humans , Clostridium Infections/microbiology , Clostridium Infections/epidemiology , Clostridium Infections/transmission , Genome, Bacterial , Drug Resistance, Bacterial/genetics , Anti-Bacterial Agents/pharmacology , Polymorphism, Single Nucleotide , Genotype
6.
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.
BMC Microbiol ; 24(1): 177, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783194

ABSTRACT

BACKGROUND: Clostridioides difficile is the main pathogen of antimicrobial-associated diarrhoea and health care facility-associated infectious diarrhoea. This study aimed to investigate the prevalence, toxin genotypes, and antibiotic resistance of C. difficile among hospitalized patients in Xi'an, China. RESULTS: We isolated and cultured 156 strains of C. difficile, representing 12.67% of the 1231 inpatient stool samples collected. Among the isolates, tcdA + B + strains were predominant, accounting for 78.2% (122/156), followed by 27 tcdA-B + strains (27/156, 17.3%) and 6 binary toxin gene-positive strains. The positive rates of three regulatory genes, tcdC, tcdR, and tcdE, were 89.1% (139/156), 96.8% (151/156), and 100%, respectively. All isolates were sensitive to metronidazole, and the resistance rates to clindamycin and cephalosporins were also high. Six strains were found to be resistant to vancomycin. CONCLUSION: Currently, the prevalence rate of C. difficile infection (CDI) in Xi'an is 12.67% (156/1231), with the major toxin genotype of the isolates being tcdA + tcdB + cdtA-/B-. Metronidazole and vancomycin were still effective drugs for the treatment of CDI, but we should pay attention to antibiotic management and epidemiological surveillance of CDI.


Subject(s)
Anti-Bacterial Agents , Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Feces , Genotype , Hospitals , Clostridioides difficile/genetics , Clostridioides difficile/drug effects , Clostridioides difficile/isolation & purification , Clostridioides difficile/classification , Humans , China/epidemiology , Anti-Bacterial Agents/pharmacology , Clostridium Infections/microbiology , Clostridium Infections/epidemiology , Bacterial Toxins/genetics , Hospitals/statistics & numerical data , Feces/microbiology , Drug Resistance, Bacterial/genetics , Prevalence , Microbial Sensitivity Tests , Female , Middle Aged , Male , Aged , Adult , Bacterial Proteins/genetics , Diarrhea/microbiology , Diarrhea/epidemiology , Metronidazole/pharmacology , Young Adult , Enterotoxins/genetics , Adolescent , Vancomycin/pharmacology , Clindamycin/pharmacology , Aged, 80 and over
11.
Mol Microbiol ; 121(6): 1182-1199, 2024 06.
Article in English | MEDLINE | ID: mdl-38690761

ABSTRACT

The dipeptide D-Ala-D-Ala is an essential component of peptidoglycan and the target of vancomycin. Most Clostridioides difficile strains possess the vanG operon responsible for the synthesis of D-Ala-D-Ser, which can replace D-Ala-D-Ala in peptidoglycan. The C. difficile vanG operon is regulated by a two-component system, VanRS, but is not induced sufficiently by vancomycin to confer resistance to this antibiotic. Surprisingly, in the absence of the VanS histidine kinase (HK), the vanG operon is still induced by vancomycin and also by another antibiotic, ramoplanin, in a VanR-dependent manner. This suggested the cross-regulation of VanR by another HK or kinases that are activated in the presence of certain lipid II-targeting antibiotics. We identified these HKs as CD35990 and CD22880. However, mutations in either or both HKs did not affect the regulation of the vanG operon in wild-type cells suggesting that intact VanS prevents the cross-activation of VanR by non-cognate HKs. Overproduction of VanR in the absence of VanS, CD35990, and CD22880 led to high expression of the vanG operon indicating that VanR can potentially utilize at least one more phosphate donor for its activation. Candidate targets of CD35990- and CD22880-mediated regulation in the presence of vancomycin or ramoplanin were identified by RNA-Seq.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Clostridioides difficile , Gene Expression Regulation, Bacterial , Histidine Kinase , Operon , Vancomycin Resistance , Vancomycin , Operon/genetics , Clostridioides difficile/genetics , Clostridioides difficile/drug effects , Clostridioides difficile/metabolism , Histidine Kinase/metabolism , Histidine Kinase/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Vancomycin/pharmacology , Vancomycin Resistance/genetics , Anti-Bacterial Agents/pharmacology , Depsipeptides/pharmacology , Transcription Factors
12.
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
13.
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
14.
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
15.
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
16.
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
17.
J Glob Antimicrob Resist ; 37: 93-99, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38552878

ABSTRACT

OBJECTIVES: Eravacycline, a new tetracycline derivative, exhibits broad-spectrum antimicrobial susceptibility. This study aimed to comprehensively investigate in vitro activities of eravacycline, tigecycline, and ertapenem against various Gram-positive, Gram-negative, and anaerobic bacteria. METHODS: Minimum inhibitory concentrations (MICs) were determined using the broth microdilution method. The following bacterial species were collected: vancomycin-sensitive (VS) Enterococci species, vancomycin-resistant Enterococci species (VRE), Staphylococcus aureus, Streptococcus anginosus, Bacteroides species, Clostridioides difficile, Clostridium innocuum, Clostridium perfringens, Parabacteroides distasonis, and Stenotrophomonas maltophilia. RESULTS: We found that eravacycline exhibited superior in vitro activity compared to tigecycline and ertapenem. Notably, it exhibited the lowest MIC90 for several bacterial species, including VS E. faecalis (0.12 µg/mL), VS E. faecium (0.12 µg/mL), and others. Besides, VRE was susceptible to eravacycline (MIC90:0.12 µg/mL) and tigecycline (MIC90:0.12 µg/mL), but was all resistant to ertapenem (MIC90 > 64 µg/mL). S. aureus was also susceptible to eravacycline (MIC90:0.5 µg/mL) as well as tigecycline (MIC90:1.0 µg/mL). Furthermore, S. anginosus showed higher susceptibility to eravacycline (MIC90:2.0 µg/mL) and tigecycline (MIC90:4.0 µg/mL), but lower to ertapenem (MIC90:32.0 µg/mL). Eravacycline and tigecycline also demonstrated good susceptibility to anaerobes, including Bacteroides species (susceptibility rate: 100%), P. distasonis (100%), C. difficile (94.1‒100%), C. innocuum (94.1‒96.1%), and C. perfringens (88.9‒96.3%). For S. maltophilia, both tigecycline and eravacycline showed an MIC90 of 2 µg/mL. A moderate-to-strong correlation (rho = 0.608-0.804, P < 0.001) was noted between the MIC values of eravacycline and tigecycline against various bacterial species. CONCLUSIONS: Our study highlights the potential of eravacycline as an effective treatment option for multidrug-resistant bacterial infections.


Subject(s)
Anti-Bacterial Agents , Bacteria, Anaerobic , Microbial Sensitivity Tests , Tetracyclines , Tigecycline , Tigecycline/pharmacology , Tetracyclines/pharmacology , Humans , Anti-Bacterial Agents/pharmacology , Bacteria, Anaerobic/drug effects , Taiwan , Ertapenem/pharmacology , Staphylococcus aureus/drug effects , Bacteria, Aerobic/drug effects , Bacteria, Aerobic/isolation & purification , Vancomycin-Resistant Enterococci/drug effects , Streptococcus anginosus/drug effects , Streptococcus anginosus/isolation & purification , Clostridioides difficile/drug effects , Stenotrophomonas maltophilia/drug effects , Vancomycin/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects
18.
Anaerobe ; 86: 102821, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38336258

ABSTRACT

BACKGROUND: Clostridioides difficile (formerly Clostridium difficile) is well-documented in Europe and North America to be a common cause of healthcare-associated gastrointestinal tract infections. In contrast, C difficile infection (CDI) is infrequently reported in literature from Asia, which may reflect a lack of clinician awareness. We conducted a narrative review to better understand CDI burden in Asia. METHODS: We searched the PubMed database for English language articles related to C difficile, Asia, epidemiology, and molecular characteristics (eg, ribotype, antimicrobial resistance). RESULTS: Fifty-eight articles that met eligibility criteria were included. C difficile prevalence ranged from 7.1% to 45.1 % of hospitalized patients with diarrhea, and toxigenic strains among all C difficile in these patients ranged from 68.2% to 91.9 % in China and from 39.0% to 60.0 % outside of China. Widespread C difficile ribotypes were RT017, RT014/020, RT012, and RT002. Recurrence in patients with CDI ranged from 3.0% to 17.2 %. Patients with CDI typically had prior antimicrobial use recently. High rates of resistance to ciprofloxacin, clindamycin, and erythromycin were frequently reported. CONCLUSION: The regional CDI burden in Asia is still incompletely documented, seemingly due to low awareness and limited laboratory testing. Despite this apparent under recognition, the current CDI burden highlights the need for broader surveillance and for application of preventative measures against CDI in Asia.


Subject(s)
Clostridioides difficile , Clostridium Infections , Humans , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , Clostridioides difficile/drug effects , Clostridioides difficile/classification , Prevalence , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Asia, Southeastern/epidemiology , Ribotyping , Drug Resistance, Bacterial , Diarrhea/microbiology , Diarrhea/epidemiology
19.
Clin Infect Dis ; 78(6): 1462-1472, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38305378

ABSTRACT

BACKGROUND: Exposure to antibiotics predisposes to dysbiosis and Clostridioides difficile infection (CDI) that can be severe, recurrent (rCDI), and life-threatening. Nonselective drugs that treat CDI and perpetuate dysbiosis are associated with rCDI, in part due to loss of microbiome-derived secondary bile acid (SBA) production. Ridinilazole is a highly selective drug designed to treat CDI and prevent rCDI. METHODS: In this phase 3 superiority trial, adults with CDI, confirmed with a stool toxin test, were randomized to receive 10 days of ridinilazole (200 mg twice daily) or vancomycin (125 mg 4 times daily). The primary endpoint was sustained clinical response (SCR), defined as clinical response and no rCDI through 30 days after end of treatment. Secondary endpoints included rCDI and change in relative abundance of SBAs. RESULTS: Ridinilazole and vancomycin achieved an SCR rate of 73% versus 70.7%, respectively, a treatment difference of 2.2% (95% CI: -4.2%, 8.6%). Ridinilazole resulted in a 53% reduction in recurrence compared with vancomycin (8.1% vs 17.3%; 95% CI: -14.1%, -4.5%; P = .0002). Subgroup analyses revealed consistent ridinilazole benefit for reduction in rCDI across subgroups. Ridinilazole preserved microbiota diversity, increased SBAs, and did not increase the resistome. Conversely, vancomycin worsened CDI-associated dysbiosis, decreased SBAs, increased Proteobacteria abundance (∼3.5-fold), and increased the resistome. CONCLUSIONS: Although ridinilazole did not meet superiority in SCR, ridinilazole greatly reduced rCDI and preserved microbiome diversity and SBAs compared with vancomycin. These findings suggest that treatment of CDI with ridinilazole results in an earlier recovery of gut microbiome health. Clinical Trials Registration.Ri-CoDIFy 1 and 2: NCT03595553 and NCT03595566.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Gastrointestinal Microbiome , Vancomycin , Humans , Vancomycin/therapeutic use , Vancomycin/adverse effects , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Male , Female , Middle Aged , Double-Blind Method , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/adverse effects , Aged , Clostridioides difficile/drug effects , Gastrointestinal Microbiome/drug effects , Adult , Treatment Outcome , Metabolome/drug effects , Oxadiazoles/therapeutic use , Oxadiazoles/adverse effects , Dysbiosis/chemically induced , Benzimidazoles , Pyridines
20.
Adv Healthc Mater ; 13(14): e2303295, 2024 06.
Article in English | MEDLINE | ID: mdl-38321619

ABSTRACT

The emerging antibiotic resistance has been named by the World Health Organization (WHO) as one of the top 10 threats to public health. Notably, methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus faecalis (VREF) are designated as serious threats, whereas Clostridioides difficile (C. difficile) is recognized as one of the most urgent threats to human health and unmet medical need. Herein, they report the design and application of novel biodegradable polymers - the lipidated antimicrobial guanidinylate polycarbonates. These polymers showed potent antimicrobial activity against a panel of bacteria with fast-killing kinetics and low resistance development tendency, mainly due to their bacterial membrane disruption mechanism. More importantly, the optimal polymer showed excellent antibacterial activity against C. difficile infection (CDI) in vivo via oral administration. In addition, compared with vancomycin, the polymer demonstrated a much-prolonged therapeutic effect and virtually diminished recurrence rate of CDI. The convenient synthesis, easy scale-up, low cost, as well as biodegradability of this class of polycarbonates, together with their in vitro broad-spectrum antimicrobial activity and orally in vivo efficacy against CDI, suggest the great potential of lipidated guandinylate polycarbonates as a new class of antibacterial biomaterials to treat CDI and combat emerging antibiotic resistance.


Subject(s)
Clostridioides difficile , Polycarboxylate Cement , Clostridioides difficile/drug effects , Animals , Polycarboxylate Cement/chemistry , Polycarboxylate Cement/pharmacology , Mice , Administration, Oral , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests , Guanidines/chemistry , Guanidines/pharmacology , Clostridium Infections/drug therapy , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...