Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
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
2.
Antimicrob Agents Chemother ; 68(3): e0162123, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38364016

ABSTRACT

Antimicrobial resistance is emerging in clinical strains of Clostridioides difficile. Ibezapolstat (IBZ) is a DNA polymerase IIIC inhibitor that has completed phase II clinical trials. IBZ has potent in vitro activity against wild-type, susceptible strains but its effect on C. difficile strains with reduced susceptibility to metronidazole (MTZ), vancomycin (VAN), or fidaxomicin (FDX) has not been tested. The primary objective of this study was to test the antibacterial properties of IBZ against multidrug-resistant C. difficile strains. The in vitro activity, bactericidal, and time-kill activity of IBZ versus comparators were evaluated against 100 clinical strains of which 59 had reduced susceptibility to other C. difficile antibiotics. Morphologic changes against a multidrug resistance strain were visualized by light and scanning electron microscopy. The overall IBZ MIC50/90 values (µg/mL) for evaluated C. difficile strains were 4/8, compared with 2/4 for VAN, 0.5/1 for FDX, and 0.25/4 for MTZ. IBZ MIC50/90 values did not differ based on non-susceptibility to antibiotic class or number of classes to which strains were non-susceptible. IBZ bactericidal activity was similar to the minimum inhibitory concentration (MIC) and maintained in wild-type and non-susceptible strains. Time-kill assays against two laboratory wild-type and two clinical non-susceptible strains demonstrated sustained IBZ activity despite reduced killing by comparator antibiotics for IBZ and VAN non-susceptible strains. Microscopy visualized increased cell lengthening and cellular damage in multidrug-resistant strains exposed to IBZ sub-MIC concentrations. This study demonstrated the potent antibacterial activity of IBZ against a large collection of C. difficile strains including multidrug-resistant strains. This study highlights the therapeutic potential of IBZ against multidrug-resistant strains of C. difficile.


Subject(s)
Anti-Infective Agents , Clostridioides difficile , Clostridium Infections , Purine Nucleosides , Humans , Clostridioides , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Infective Agents/pharmacology , Vancomycin/pharmacology , Vancomycin/therapeutic use , Metronidazole/pharmacology , Metronidazole/therapeutic use , Fidaxomicin/pharmacology , Fidaxomicin/therapeutic use , Microbial Sensitivity Tests
3.
Antimicrob Agents Chemother ; 68(3): e0122223, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38265216

ABSTRACT

Clostridioides difficile infection (CDI) is a leading cause of hospital-acquired diarrhea, which often stems from disruption of the gut microbiota by broad-spectrum antibiotics. The increasing prevalence of antibiotic-resistant C. difficile strains, combined with disappointing clinical trial results for recent antibiotic candidates, underscores the urgent need for novel CDI antibiotics. To this end, we investigated C. difficile enoyl ACP reductase (CdFabK), a crucial enzyme in de novo fatty acid synthesis, as a drug target for microbiome-sparing antibiotics. To test this concept, we evaluated the efficacy and in vivo spectrum of activity of the phenylimidazole analog 296, which is validated to inhibit intracellular CdFabK. Against major CDI-associated ribotypes 296 had an Minimum inhibitory concentration (MIC90) of 2 µg/mL, which was comparable to vancomycin (1 µg/mL), a standard of care antibiotic. In addition, 296 achieved high colonic concentrations and displayed dosed-dependent efficacy in mice with colitis CDI. Mice that were given 296 retained colonization resistance to C. difficile and had microbiomes that resembled the untreated mice. Conversely, both vancomycin and fidaxomicin induced significant changes to mice microbiomes, in a manner consistent with prior reports. CdFabK, therefore, represents a potential target for microbiome-sparing CDI antibiotics, with phenylimidazoles providing a good chemical starting point for designing such agents.


Subject(s)
Clostridioides difficile , Clostridium Infections , Animals , Mice , Vancomycin/pharmacology , Oxidoreductases , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Fidaxomicin/pharmacology , Clostridium Infections/drug therapy
4.
BMC Vet Res ; 19(1): 238, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37974163

ABSTRACT

BACKGROUND: Previous studies have demonstrated that fidaxomicin, a macrocyclic lactone antibiotic used to treat recurrent Clostridioides difficile-associated diarrhea, also displays potent in vitro bactericidal activity against Clostridium perfringens strains isolated from humans. However, to date, there is no data on the susceptibility to fidaxomicin of C. perfringens strains of animal origin. On the other hand, although combination therapy has become popular in human and veterinary medicine, limited data are available on the effects of antibiotic combinations on C. perfringens. We studied the in vitro response of 21 C. perfringens strains obtained from dogs and cats to fidaxomicin and combinations of fidaxomicin with six other antibiotics. RESULTS: When tested by an agar dilution method, fidaxomicin minimum inhibitory concentrations (MICs) ranged between 0.004 and 0.032 µg/ml. Moreover, the results of Etest-based combination assays revealed that the incorporation of fidaxomicin into the test medium at a concentration equivalent to half the MIC significantly increased the susceptibility of isolates to metronidazole and erythromycin in 71.4% and 61.9% of the strains, respectively, and the susceptibility to clindamycin, imipenem, levofloxacin, and vancomycin in 42.9-52.4% of the strains. In contrast, » × MIC concentrations of fidaxomicin did not have any effect on levofloxacin and vancomycin MICs and only enhanced the effects of clindamycin, erythromycin, imipenem, and metronidazole in ≤ 23.8% of the tested strains. CONCLUSIONS: The results of this study demonstrate that fidaxomicin is highly effective against C. perfringens strains of canine and feline origin. Although fidaxomicin is currently considered a critically important antimicrobial that has not yet been licensed for veterinary use, we consider that the results reported in this paper provide useful baseline data to track the possible emergence of fidaxomicin resistant strains of C. perfringens in the veterinary setting.


Subject(s)
Cat Diseases , Clostridioides difficile , Clostridium Infections , Dog Diseases , Cats , Animals , Dogs , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Fidaxomicin/pharmacology , Clostridium perfringens , Cat Diseases/drug therapy , Vancomycin/pharmacology , Metronidazole/pharmacology , Clindamycin , Levofloxacin/pharmacology , Clostridium Infections/drug therapy , Clostridium Infections/veterinary , Dog Diseases/drug therapy , Imipenem/pharmacology , Erythromycin/pharmacology , Diarrhea/drug therapy , Diarrhea/veterinary , Microbial Sensitivity Tests/veterinary
5.
Clin Ther ; 45(4): 356-362, 2023 04.
Article in English | MEDLINE | ID: mdl-36906440

ABSTRACT

PURPOSE: Bezlotoxumab is approved for prevention of recurrent Clostridioides (Clostridium) difficile infection (CDI) in adults receiving antibacterial treatment for CDI who are at high risk for recurrent CDI. Previous studies have shown that although serum albumin levels are an important predictor for bezlotoxumab exposure, this has no clinically meaningful impact on efficacy. This pharmacokinetic modeling study assessed whether hematopoietic stem cell transplant (HSCT) recipients, at increased risk of CDI and exhibiting decreased albumin levels within the first month posttransplant, are at risk of clinically relevant reductions in bezlotoxumab exposure. METHODS: Observed bezlotoxumab concentration-time data pooled from participants in Phase III trials MODIFY I and II (ClinicalTrials.gov identifiers NCT01241552/NCT01513239) and three Phase I studies (PN004, PN005, and PN006) were used to predict bezlotoxumab exposures in two adult post-HSCT populations: A Phase Ib study of posaconazole including allogeneic HSCT recipients (ClinicalTrials.gov identifier NCT01777763; posaconazole-HSCT population); and a Phase III study of fidaxomicin for CDI prophylaxis (ClinicalTrials.gov identifier NCT01691248; fidaxomicin-HSCT population). The bezlotoxumab PK model used the minimum albumin level for each individual in post-HSCT populations to mimic a "worst-case scenario." FINDINGS: Predicted worst-case bezlotoxumab exposures for the posaconazole-HSCT population (N = 87) were decreased by 10.8% versus bezlotoxumab exposures observed in the pooled Phase III/Phase I data set (N = 1587). No further decrease was predicted for the fidaxomicin-HSCT population (N = 350). IMPLICATIONS: Based on published population pharmacokinetic data, the predicted decrease in bezlotoxumab exposure in the post-HSCT populations is not expected to have a clinically meaningful effect on bezlotoxumab efficacy at the recommended 10 mg/kg dose. Dose modification is therefore not required in the hypoalbuminemia setting expected post-HSCT.


Subject(s)
Clostridioides difficile , Clostridium Infections , Hematopoietic Stem Cell Transplantation , Adult , Humans , Fidaxomicin/pharmacology , Fidaxomicin/therapeutic use , Anti-Bacterial Agents/adverse effects , Clostridium Infections/drug therapy , Clostridium Infections/epidemiology , Clostridium Infections/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Albumins/therapeutic use
6.
J Med Microbiol ; 71(6)2022 Jun.
Article in English | MEDLINE | ID: mdl-35708979

ABSTRACT

Introduction. Nontuberculous mycobacteria (NTM) infections are increasing worldwide and are relatively resistant to many of the first- and second-line drugs to treat tuberculosis. Macrolide antibiotics, such as clarithromycin and azithromycin, are the key drugs for treating NTM infections. Fidaxomicin is a macrolide antibiotic that is widely used in treating Clostridium difficle (C.difficile) infections, and has high in vitro activity against Mycobacterium tuberculosis especially multidrug-resistant tuberculosis (MDR-TB) and has no cross-resistance with rifampicin.Hypothesis. Fidaxomicin may have in vitro activity against NTM strains.Aim. To find that whether the macrolide antibiotic fidaxomicin has in vitro activity against NTM strains.Methodology. Fidaxomicin used in this study was firstly tested on C. difficile reference strains and has shown to be effective and workable. And then 28 rapidly growing mycobacteria (RGM), 12 slowly growing mycobacteria (SGM) reference strains and 103 NTM clinical isolates were tested by the microplate-based AlamarBlue assay (MABA) method to determine the MICs. Fidaxomicin, rifampicin and clarithromycin were tested against M. abcessus complex subspecies 14 M. abscessus and 5 M. massiliense strains for inducible resistance determination.Results. In total, 21 out of 28 RGM and 9 of 12 SGM reference strains have the MICs of fidaxomicin at or below 1 µg ml-1. Fidaxomicin also showed low MIC values for some clinical isolates including M. abscessus complex, M. avium complex, M. fortuitum, M. kansasii and M. parascrofulaceum. Fidaxomicin also has no inducible macrolide resistance in M. abscessus complex in comparison with clarithromycin.Conclusion. Fidaxomicin has high in vitro activity against most of the NTM reference strains and some prevalent NTM clinical isolates. This promising finding warrants further investigation on the actions of fidaxomicn in vivo and as a potential antibiotic for NTM treatment.


Subject(s)
Clostridioides difficile , Mycobacterium Infections, Nontuberculous , Mycobacterium , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Clarithromycin/pharmacology , Clarithromycin/therapeutic use , Drug Resistance, Bacterial , Fidaxomicin/pharmacology , Humans , Macrolides/pharmacology , Microbial Sensitivity Tests , Mycobacterium Infections, Nontuberculous/microbiology , Nontuberculous Mycobacteria , Rifampin/pharmacology , Rifampin/therapeutic use
7.
Nature ; 604(7906): 541-545, 2022 04.
Article in English | MEDLINE | ID: mdl-35388215

ABSTRACT

Fidaxomicin (Fdx) is widely used to treat Clostridioides difficile (Cdiff) infections, but the molecular basis of its narrow-spectrum activity in the human gut microbiome remains unknown. Cdiff infections are a leading cause of nosocomial deaths1. Fidaxomicin, which inhibits RNA polymerase, targets Cdiff with minimal effects on gut commensals, reducing recurrence of Cdiff infection2,3. Here we present the cryo-electron microscopy structure of Cdiff RNA polymerase in complex with fidaxomicin and identify a crucial fidaxomicin-binding determinant of Cdiff RNA polymerase that is absent in most gut microbiota such as Proteobacteria and Bacteroidetes. By combining structural, biochemical, genetic and bioinformatic analyses, we establish that a single residue in Cdiff RNA polymerase is a sensitizing element for fidaxomicin narrow-spectrum activity. Our results provide a blueprint for targeted drug design against an important human pathogen.


Subject(s)
Clostridioides difficile , Clostridium Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Clostridioides , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Cryoelectron Microscopy , DNA-Directed RNA Polymerases , Fidaxomicin/chemistry , Fidaxomicin/pharmacology , Fidaxomicin/therapeutic use , Humans
8.
J Biol Chem ; 298(4): 101752, 2022 04.
Article in English | MEDLINE | ID: mdl-35189142

ABSTRACT

RNA polymerase (RNAP) binding protein A (RbpA) is essential for mycobacterial viability and regulates transcription initiation by increasing the stability of the RNAP-promoter open complex (RPo). RbpA consists of four domains: an N-terminal tail (NTT), a core domain (CD), a basic linker, and a sigma interaction domain. We have previously shown that truncation of the RbpA NTT and CD increases RPo stabilization by RbpA, implying that these domains inhibit this activity of RbpA. Previously published structural studies showed that the NTT and CD are positioned near multiple RNAP-σA holoenzyme functional domains and predict that the RbpA NTT contributes specific amino acids to the binding site of the antibiotic fidaxomicin (Fdx), which inhibits the formation of the RPo complex. Furthermore, deletion of the NTT results in decreased Mycobacterium smegmatis sensitivity to Fdx, but whether this is caused by a loss in Fdx binding is unknown. We generated a panel of rbpA mutants and found that the RbpA NTT residues predicted to directly interact with Fdx are partially responsible for RbpA-dependent Fdx activity in vitro, while multiple additional RbpA domains contribute to Fdx activity in vivo. Specifically, our results suggest that the RPo-stabilizing activity of RbpA decreases Fdx activity in vivo. In support of the association between RPo stability and Fdx activity, we find that another factor that promotes RPo stability in bacteria, CarD, also impacts to Fdx sensitivity. Our findings highlight how RbpA and other factors may influence RNAP dynamics to affect Fdx sensitivity.


Subject(s)
Fidaxomicin , Mycobacterium smegmatis , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Fidaxomicin/pharmacology , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/genetics , Promoter Regions, Genetic , Sigma Factor/metabolism
9.
Microb Drug Resist ; 27(12): 1672-1676, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34037477

ABSTRACT

In this study, we report the results of the epidemiological analysis of Clostridioides difficile ribotypes (RTs) and antimicrobial susceptibility testing. Most isolates were RT027, representing 73% (84/115) of isolates. No isolates with reduced susceptibility to fidaxomicin were found; however, 38 (33.04%) isolates had reduced susceptibility to metronidazole, and 7 isolates (6.1%) had reduced susceptibility to vancomycin. These findings highlight the need for continuous surveillance of C. difficile RTs and antimicrobial susceptibility testing.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Drug Resistance, Bacterial/genetics , Fidaxomicin/pharmacology , Genes, Bacterial , Humans , Metronidazole/pharmacology , Mexico , Microbial Sensitivity Tests , Ribotyping , Vancomycin/pharmacology
10.
J Med Microbiol ; 70(3)2021 Mar.
Article in English | MEDLINE | ID: mdl-33593474

ABSTRACT

This study aimed to evaluate whether the antibiotic fidaxomicin has in vitro activity against Mycobacterium tuberculosis (Mtb). 38 fully drug-sensitive Mtb strains and 34 multidrug-resistant tuberculosis (MDR-TB) strains were tested using the microplate alamar blue assay (MABA) method to determine the minimum inhibitory concentrations (MICs) for fidaxomicin and rifampicin. Fidaxomicin has high in vitro activity against Mtb and is a potential drug to treat Mtb, and MDR-TB infections in particular.


Subject(s)
Antitubercular Agents/pharmacology , Fidaxomicin/pharmacology , Mycobacterium tuberculosis/drug effects , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Microbial Sensitivity Tests , Mutation , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/isolation & purification , Rifampin/pharmacology , Tuberculosis/microbiology , Tuberculosis, Multidrug-Resistant/microbiology
11.
Int J Infect Dis ; 103: 226-233, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33188906

ABSTRACT

PURPOSE: We aimed to evaluate the efficacy of different antibiotic regimens for the treatment of Clostridioides difficile infection (CDI) with regard to the CDI episode number and disease severity. METHODS: An observation cohort study included 271 CDI patients hospitalised between 2013-2016. Univariate logistic regression was used to evaluate the association between patients' clinical outcome (sustained clinical cure or recurrence) in a 60-day follow-up and the antibiotic regimen used (oral metronidazole, oral vancomycin, combination of oral vancomycin and metronidazole, oral fidaxomicin). Subgroup analyses, based on CDI episode number and severity, were performed. RESULTS: In the overall population, fidaxomicin was superior to metronidazole, vancomycin or their combination, for a sustained clinical response and in the prevention of recurrent CDI (rCDI). In the subgroup analyses, fidaxomicin was superior to vancomycin or metronidazole for a sustained clinical response and in the prevention of rCDI in the initial episode, first recurrence and non-severe cases. In the oral treatment of severe CDI, fidaxomicin had a similar treatment outcome to vancomycin and none of the antibiotic treatments were superior in the prevention of rCDI. Fidaxomicin, vancomycin, or a combination of metronidazole and vancomycin, had similar outcomes for sustained clinical response and prevention of rCDI in patients with multiple rCDI. CONCLUSION: Fidaxomicin was superior to metronidazole or vancomycin for the treatment of the initial episode, first recurrence, and non-severe CDI.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Fidaxomicin/pharmacology , Metronidazole/pharmacology , Vancomycin/pharmacology , Administration, Oral , Aged , Clostridium Infections/microbiology , Cohort Studies , Female , Hospitalization , Humans , Logistic Models , Male , Middle Aged , Recurrence , Treatment Outcome
12.
Biomed Res ; 41(6): 259-268, 2020.
Article in English | MEDLINE | ID: mdl-33268670

ABSTRACT

Recently, opportunistic nosocomial infections caused by Acinetobacter baumannii have become increasingly prevalent worldwide. The pathogen often establishes biofilms that adhere to medical devices, causing chronic infections refractory to antimicrobial therapy. Clinical reports have indicated that some macrolide antibiotics are effective against chronic biofilm-related infections. In this study, we examined the direct anti-biofilm effects of seven macrolides (azithromycin, clarithromycin, erythromycin, josamycin, spiramycin, fidaxomicin, and ivermectin) on A. baumannii using a simple and newly established in vitro assay system for the swift and serial spectrophotometric determinations of two biofilm-amount indexes of viability and biomass. These macrolides were found to possess direct anti-biofilm effects exerting specific anti-biofilm effects not exclusively depending on their bacteriostatic/bactericidal effects. The anti-biofilm effect of azithromycin was found to be the strongest, while those of fidaxomicin and ivermectin were weak and limited. These results provide insights into possible adjunctive chemotherapy with macrolides for A. baumannii infection. Common five macrolides also interfered with the Agrobacterium tumefaciens NTL(pCF218) (pCF372) bioassay system of N-acyl homoserine lactones, providing insights into sample preparation for the bioassay, and putatively suggesting the actions of macrolides as remote signals in bacterial quorum sensing systems.


Subject(s)
Acinetobacter baumannii/drug effects , Acyl-Butyrolactones/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Quorum Sensing/drug effects , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Acinetobacter baumannii/growth & development , Acinetobacter baumannii/metabolism , Acyl-Butyrolactones/metabolism , Agrobacterium tumefaciens/drug effects , Agrobacterium tumefaciens/growth & development , Agrobacterium tumefaciens/metabolism , Azithromycin/pharmacology , Biofilms/growth & development , Biological Assay , Clarithromycin/pharmacology , Erythromycin/pharmacology , Fidaxomicin/pharmacology , Humans , Ivermectin/pharmacology , Josamycin/pharmacology , Microbial Sensitivity Tests , Microbial Viability/drug effects , Spiramycin/pharmacology
13.
Int J Biol Macromol ; 163: 1787-1797, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32950529

ABSTRACT

The pandemic prevalence of COVID-19 has become a very serious global health issue. Scientists all over the world have been seriously attempting in the discovery of a drug to combat SARS-CoV-2. It has been found that RNA-dependent RNA polymerase (RdRp) plays a crucial role in SARS-CoV-2 replication, and thus could be a potential drug target. Here, comprehensive computational approaches including drug repurposing and molecular docking were employed to predict an effective drug candidate targeting RdRp of SARS-CoV-2. This study revealed that Rifabutin, Rifapentine, Fidaxomicin, 7-methyl-guanosine-5'-triphosphate-5'-guanosine and Ivermectin have a potential inhibitory interaction with RdRp of SARS-CoV-2 and could be effective drugs for COVID-19. In addition, virtual screening of the compounds from ZINC database also allowed the prediction of two compounds (ZINC09128258 and ZINC09883305) with pharmacophore features that interact effectively with RdRp of SARS-CoV-2, indicating their potentiality as effective inhibitors of the enzyme. Furthermore, ADME analysis along with analysis of toxicity was also undertaken to check the pharmacokinetics and drug-likeness properties of the two compounds. Comparative structural analysis of protein-inhibitor complexes revealed that the amino acids Y32, K47, Y122, Y129, H133, N138, D140, T141, S709 and N781 are crucial for drug surface hotspot in the RdRp of SARS-CoV-2.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Drug Repositioning , Pneumonia, Viral/drug therapy , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Betacoronavirus/enzymology , COVID-19 , Coronavirus Infections/virology , Fidaxomicin/chemistry , Fidaxomicin/pharmacology , Humans , Ivermectin/chemistry , Ivermectin/pharmacology , Molecular Docking Simulation , Pandemics , Pneumonia, Viral/virology , Rifabutin/chemistry , Rifabutin/pharmacology , Rifampin/analogs & derivatives , Rifampin/chemistry , Rifampin/pharmacology , SARS-CoV-2 , Virus Replication/drug effects , COVID-19 Drug Treatment
14.
BMC Med ; 18(1): 204, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32731873

ABSTRACT

BACKGROUND: Zika virus (ZIKV) infection is a global health problem, and its complications, including congenital Zika syndrome and Guillain-Barré syndrome, constitute a continued threat to humans. Unfortunately, effective therapeutics against ZIKV infection are not available thus far. METHODS: We screened the compounds collection consisting of 1789 FDA-approved drugs by a computational docking method to obtain anti-ZIKV candidate compounds targeting ZIKV RNA-dependent RNA polymerase (RdRp). SPR (BIAcore) assay was employed to demonstrate the candidate compounds' direct binding to ZIKV RdRp, and polymerase activity assay was used to determine the inhibitory effect on ZIKV RdRp-catalyzed RNA synthesis. The antiviral effects on ZIKV in vitro and in vivo were detected in infected cultured cells and in Ifnar1-/- mice infected by ZIKV virus using plaque assay, western blotting, tissue immunofluorescence, and immunohistochemistry. RESULTS: Here, we report that a first-in-class macrocyclic antibiotic, which has been clinically used to treat Clostridium difficile infection, fidaxomicin, potently inhibits ZIKV replication in vitro and in vivo. Our data showed that fidaxomicin was effective against African and Asian lineage ZIKV in a wide variety of cell lines of various tissue origins, and prominently suppressed ZIKV infection and significantly improved survival of infected mice. In addition, fidaxomicin treatment reduced the virus load in the brains and testes, and alleviated ZIKV-associated pathological damages, such as paralysis, hunching, and neuronal necrosis in the cerebra. Furthermore, our mechanistic study showed that fidaxomicin directly bound ZIKV NS5 protein and inhibited the RNA synthesis-catalyzing activity of ZIKV RdRp. CONCLUSIONS: Our data suggest that fidaxomicin may represent an effective anti-ZIKV agent. In the light that fidaxomicin is already a clinically used drug, there might be a promising prospect in the development of fidaxomicin to be an antiviral therapeutic.


Subject(s)
Fidaxomicin/therapeutic use , RNA-Dependent RNA Polymerase/therapeutic use , Zika Virus Infection/drug therapy , Animals , Cell Line, Tumor , Chlorocebus aethiops , Female , Fidaxomicin/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , RNA-Dependent RNA Polymerase/pharmacology , Vero Cells , Zika Virus Infection/pathology
15.
Clin Transl Gastroenterol ; 11(7): e00189, 2020 07.
Article in English | MEDLINE | ID: mdl-32675703

ABSTRACT

INTRODUCTION: Clostridioides difficile infection (CDI) is common in patients with cirrhosis and is associated with poor outcomes. CDI risk factors in this population have been well characterized; however, risk factors of recurrent CDI (R-CDI) after treatment have not been explored. We sought to estimate the incidence of R-CDI and its associated risk factors in patients with cirrhosis. METHODS: We performed a cohort study of patients with cirrhosis hospitalized with CDI between 2012 and 2016. We collected patient characteristics, including detailed information on the CDI, features of the underlying liver disease, and outcomes including R-CDI, hospital readmission, and mortality. R-CDI was defined as CDI occurring 2-8 weeks after the initial episode. Cox proportional hazards model was used to identify variables independently associated with the outcomes. RESULTS: A total of 257 hospitalized patients with cirrhosis and CDI were included. CDI was community associated in 22.6%. The incidence of R-CDI was 11.9%. R-CDI was not significantly associated with medications at hospital admission or discharge. Independent risk factors of R-CDI included increased Charlson Comorbidity Index (hazard ratio [HR] 1.30; 95% confidence interval [CI]: 1.09-1.55) and use of lactulose (HR 2.58; 95% CI: 1.09-6.09). The 30-day readmission rate was 37%, and readmission was associated with increased Charlson Comorbidity Index (HR 1.12; 95% CI: 1.03-1.23) and Model for End-Stage Liver Disease score (HR 1.04; 95% CI: 1.01-1.07). The 90-day mortality was 22.8%. DISCUSSION: In patients with cirrhosis, R-CDI is associated with comorbidity burden and lactulose use. Attention to these factors might aid clinicians in efforts to prevent R-CDI and improve outcomes in this population.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides/isolation & purification , Clostridium Infections/epidemiology , Liver Cirrhosis/complications , Patient Readmission/statistics & numerical data , Aged , Anti-Bacterial Agents/therapeutic use , Clostridium Infections/diagnosis , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Comorbidity , Diagnostic Errors , Female , Fidaxomicin/pharmacology , Fidaxomicin/therapeutic use , Humans , Incidence , Lactulose/adverse effects , Laxatives , Liver Cirrhosis/diagnosis , Liver Cirrhosis/drug therapy , Male , Metronidazole/pharmacology , Metronidazole/therapeutic use , Middle Aged , Recurrence , Retrospective Studies , Risk Factors , Severity of Illness Index , Treatment Failure , Vancomycin/pharmacology , Vancomycin/therapeutic use
16.
Sci Rep ; 10(1): 7701, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32382070

ABSTRACT

Clostridioides difficile is the leading cause of nosocomial infections and a worldwide urgent public health threat. Without doubt, there is an urgent need for new effective anticlostridial agents due to the increasing incidence and severity of C. difficile infection (CDI). The aim of the present study is to investigate the in vivo efficacy of auranofin (rheumatoid arthritis FDA-approved drug) in a CDI mouse model and establish an adequate dosage for treatment. The effects of increased C. difficile inoculum, and pre-exposure to simulated gastric intestinal fluid (SGF) and simulated intestinal fluid (SIF), on the antibacterial activity of auranofin were investigated. Auranofin's in vitro antibacterial activity was stable in the presence of high bacterial inoculum size compared to vancomycin and fidaxomicin. Moreover, it maintained its anti-C. difficile activity after being exposed to SGF and SIF. Upon testing in a CDI mouse model, auranofin at low clinically achievable doses (0.125 mg/kg and 0.25 mg/kg) significantly protected mice against CDI with 100% and 80% survival, respectively. Most importantly, auranofin (0.125 mg/kg and 0.25 mg/kg) significantly prevented CDI recurrence when compared with vancomycin. Collectively, these results indicate that auranofin could potentially provide an effective, safe and quick supplement to the current approaches for treating CDI.


Subject(s)
Auranofin/pharmacology , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Cross Infection/drug therapy , Animals , Anti-Bacterial Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Clostridioides difficile/pathogenicity , Clostridium Infections/microbiology , Clostridium Infections/pathology , Cross Infection/microbiology , Cross Infection/pathology , Drug Repositioning , Fidaxomicin/pharmacology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Humans , Mice , Microbial Sensitivity Tests , Recurrence , Vancomycin/pharmacology
17.
J Infect Chemother ; 26(7): 685-692, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32224190

ABSTRACT

Clostridioides difficile infection results from a disturbance of the normal microbial flora of the colon, allowing proliferation of C. difficile and toxin production by toxigenic strains. Fidaxomicin, a macrocyclic antibiotic that prevents RNA synthesis in C. difficile and inhibits spore formation, toxin production, and cell proliferation, is clinically effective in treating C. difficile infection. As recent studies have suggested that biofilm formation influences C. difficile colonization and infection in the colon, we undertook the present study to determine the effects of fidaxomicin on C. difficile biofilm formation. Sub-minimum inhibitory concentrations (MICs) of fidaxomicin inhibited biofilm formation by C. difficile UK027 and delayed planktonic growth. Sub-MICs of vancomycin did not inhibit biofilm formation or affect planktonic growth. In C. difficile UK027 exposed to sub-MICs of fidaxomicin, mRNA expression of biofilm-related flagellin gene fliC was slightly increased compared with that of other biofilm-related genes (pilA1, cwp84, luxS, dccA, and spo0A). In conclusion, this study indicates that sub-MICs of fidaxomicin inhibit C. difficile UK027 biofilm formation by influencing cell growth and fliC transcription.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Fidaxomicin/pharmacology , Anti-Bacterial Agents/therapeutic use , Clostridioides difficile/genetics , Clostridium Infections/microbiology , Fidaxomicin/therapeutic use , Flagellin/genetics , Gene Expression Regulation, Bacterial/drug effects , Humans , Microbial Sensitivity Tests , RNA, Bacterial/biosynthesis , RNA, Bacterial/isolation & purification , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Transcription, Genetic/drug effects
18.
J Infect Chemother ; 26(5): 483-491, 2020 May.
Article in English | MEDLINE | ID: mdl-32165071

ABSTRACT

OBJECTIVE: To investigate the effect of vancomycin and fidaxomicin on the diversity of intestinal microbiota in a mouse model of Clostridioides difficile infection. METHODS: Mice were divided into 11 models (4 mice per model): 6 uninoculated models and 5 models inoculated with C. difficile BI/NAP1/027. Inoculated models were prepared using intraperitoneal clindamycin followed by inoculation with C. difficile BI/NAP1/027. Uninoculated and C. difficile-inoculated mice received 2 or 7 days' vancomycin or fidaxomicin. Clostridium butyricum MIYAIRI 588 probiotic and lactoferrin prebiotic were administered for 10 days to uninoculated mice. Intestinal microbiome composition was investigated by sequence analyses of bacterial 16S rRNA genes from faeces, and microbiota diversity estimated. RESULTS: In uninoculated, untreated ('normal') mice, Clostridia (57.8%) and Bacteroidia (32.4%) accounted for the largest proportions of gut microbiota. The proportion of Clostridia was numerically reduced in C. difficile-inoculated versus normal mice. Administration of vancomycin to C. difficile-inoculated mice reduced the proportions of Bacteroidia and Clostridia, and increased that of Proteobacteria. Administration of fidaxomicin to C. difficile-inoculated mice reduced the proportion of Clostridia to a lesser extent, but increased that of Bacteroidia. Microbiota diversity was lower in C. difficile-inoculated versus normal mice (164.5 versus 349.1 operational taxonomic units (OTUs), respectively); treatment of C. difficile-inoculated mice with 7 days' vancomycin reduced diversity to a greater extent than did 7 days' fidaxomicin treatment (26.2 versus 134.2 OTUs, respectively). CONCLUSIONS: Both C. difficile inoculation and treatment with vancomycin or fidaxomicin reduced microbiota diversity; however, dysbiosis associated with fidaxomicin was milder than with vancomycin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Fidaxomicin/pharmacology , Gastrointestinal Microbiome/drug effects , Vancomycin/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Clostridioides difficile/isolation & purification , Clostridium Infections/microbiology , Disease Models, Animal , Feces/microbiology , Fidaxomicin/therapeutic use , Humans , Mice , Microbiota/drug effects , RNA, Ribosomal, 16S/genetics , Vancomycin/therapeutic use
19.
Nihon Yakurigaku Zasshi ; 154(4): 217-229, 2019.
Article in Japanese | MEDLINE | ID: mdl-31597902

ABSTRACT

Fidaxomicin (Dafclir® Tablets 200 mg) is a member of a novel class of oral, 18-membered macrocyclic antibiotic agents used for the treatment of patients with Clostridioides (Clostridium) difficile (C. difficile) infection (CDI), approved in Japan in July 2018. Preclinical studies demonstrated that fidaxomicin inhibits RNA synthesis by bacterial DNA-dependent RNA polymerase derived from C. difficile, shows antibacterial activities against C. difficile clinically isolated in Japan, and is bactericidal against C. difficile. Fidaxomicin was less likely to disrupt gut microflora due to its narrow antimicrobial spectrum, showing antibacterial activities against limited gram-positive bacteria including C. difficile, but not against gram-negative bacteria, as determined by minimum inhibitory concentration (MIC) measurement against American Type Culture Collection strains. Fidaxomicin inhibited spore production, subsequent spore recovery/outgrowth after removal of fidaxomicin, outgrowth to vegetative cells, and toxin production under fidaxomicin at lower MIC. Additionally, it had protective effects on lethal CDI in animal models. In clinical studies conducted in Europe, US, and Japan, fidaxomicin 200 mg twice daily for 10 days showed higher clinical cure, higher global cure (cure with no recurrence), and lower recurrence rate compared with oral vancomycin 125 mg four times daily for 10 days. Adverse events observed in the fidaxomicin group were similar to those in the vancomycin group, and no clinically important findings regarding safety and tolerability were reported. In conclusion, in vitro, in vivo and clinical evidence indicate that fidaxomicin is an effective treatment for C. difficile, with limited disruption to gut microflora, for adult patients with CDI in Japan.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Fidaxomicin/pharmacology , Animals , Clinical Trials as Topic , Humans , Japan , Microbial Sensitivity Tests
20.
Article in English | MEDLINE | ID: mdl-31085514

ABSTRACT

In 2011, we initiated a sentinel surveillance network to assess changes in Clostridioides (formerly Clostridium) difficile antimicrobial susceptibility to fidaxomicin from 6 geographically dispersed medical centers in the United States. This report summarizes data from 2013 to 2016. C. difficile isolates or toxin-positive stools from patients were referred to a central laboratory. Antimicrobial susceptibility was determined by agar dilution. CLSI, EUCAST, or FDA breakpoints were used, where applicable. Toxin gene profiles were characterized by multiplex PCR on each isolate. A random sample of approximately 40% of isolates, stratified by institution and year, was typed by restriction endonuclease analysis (REA). Among 1,889 isolates from 2013 to 2016, the fidaxomicin MIC90 was 0.5 µg/ml; all isolates were inhibited at ≤1 µg/ml. There were decreases in metronidazole and vancomycin MICs over time. Clindamycin resistance remained unchanged (27.3%). An increase in imipenem resistance was observed. By 2015 to 2016, moxifloxacin resistance decreased in all centers. The proportion of BI isolates decreased from 25.5% in 2011 to 2012 to 12.8% in 2015 to 2016 (P < 0.001). The BI REA group correlated with moxifloxacin resistance (BI 84% resistant versus non-BI 12.5% resistant). Fidaxomicin MICs have not changed among C. difficile isolates of U.S. origin over 5 years post licensure. There has been an overall decrease in MICs for vancomycin, metronidazole, moxifloxacin, and rifampin and an increase in isolates resistant to imipenem. Moxifloxacin resistance remained high among the BI REA group, but the proportion of BI isolates has decreased. Continued geographic variations in REA groups and antimicrobial resistance persist.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridium Infections/microbiology , Diarrhea/microbiology , Fidaxomicin/pharmacology , ADP Ribose Transferases/genetics , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Clindamycin/pharmacology , Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Enterotoxins/genetics , Humans , Imipenem/pharmacology , Microbial Sensitivity Tests , Prohibitins , Sentinel Surveillance , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...