Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Sci Rep ; 13(1): 14733, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37679389

ABSTRACT

Clostridioides difficile infections (CDIs) are responsible for a significant number of antibiotic-associated diarrheal cases. The standard-of-care antibiotics for C. difficile are limited to fidaxomicin and vancomycin, with the recently obsolete metronidazole recommended if both are unavailable. No new antimicrobials have been approved for CDI since fidaxomicin in 2011, despite varying rates of treatment failure among all standard-of-care drugs. Drug repurposing is a rational strategy to generate new antimicrobials out of existing therapeutics approved for other indications. Auranofin is a gold-containing anti-rheumatic drug with antimicrobial activity against C. difficile and other microbes. In a previous report, our group hypothesized that inhibition of selenoprotein biosynthesis was auranofin's primary mechanism of action against C. difficile. However, in this study, we discovered that C. difficile mutants lacking selenoproteins are still just as sensitive to auranofin as their respective wild-type strains. Moreover, we found that selenite supplementation dampens the activity of auranofin against C. difficile regardless of the presence of selenoproteins, suggesting that selenite's neutralization of auranofin is not because of compensation for a chemically induced selenium deficiency. Our results clarify the findings of our original study and may aid drug repurposing efforts in discovering the compound's true mechanism of action against C. difficile.


Subject(s)
Auranofin , Clostridioides difficile , Auranofin/pharmacology , Clostridioides , Fidaxomicin , Selenious Acid , Selenoproteins/genetics
2.
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
3.
Eur J Clin Microbiol Infect Dis ; 38(6): 1105-1111, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30989419

ABSTRACT

The cost of treating Clostridium difficile infection (CDI) in Spain is substantial. Findings from the randomised, controlled, open-label, phase 3b/4 EXTEND study showed that an extended-pulsed fidaxomicin (EPFX) regimen was associated with improved sustained clinical cure and reduced recurrence of CDI versus vancomycin in patients aged 60 years and older. We assessed the cost-effectiveness of EPFX versus vancomycin for the treatment of CDI in patients aged 60 years and older from the perspective of the National Health System (NHS) in Spain. We used a Markov model with six health states and 1-year time horizon. Health resources, their unit costs and utilities were based on published sources. Key efficacy data and transition probabilities were obtained from the EXTEND study and published sources. A panel of Spanish clinical experts validated all model assumptions. In the analysis, 0.638 and 0.594 quality-adjusted life years (QALYs) per patient were obtained with EPFX and vancomycin, respectively, with a gain of 0.044 QALYs with EPFX. The cost per patient treated with EPFX and vancomycin was estimated to be €10,046 and €10,693, respectively, with a saving of €647 per patient treated with EPFX. For willingness-to-pay thresholds of €20,000, €25,000 and €30,000 per QALY gained, the probability that EPFX was the most cost-effective treatment was 99.3%, 99.5% and 99.9%, respectively. According to our economic model and the assumptions based on the Spanish NHS, EPFX is cost-effective compared with vancomycin for the first-line treatment of CDI in patients aged 60 years and older.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Clostridium Infections/drug therapy , Cost-Benefit Analysis , Fidaxomicin/administration & dosage , Vancomycin/administration & dosage , Aged , Aged, 80 and over , Anti-Bacterial Agents/economics , Clostridioides difficile , Clostridium Infections/economics , Health Care Costs , Humans , Middle Aged , Models, Economic , National Health Programs , Quality-Adjusted Life Years , Spain , Treatment Outcome
4.
J Antimicrob Chemother ; 73(1): 102-108, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29029221

ABSTRACT

BACKGROUND: Clostridium difficile, a spore-forming Gram-positive anaerobic bacillus, is the most common causative agent of healthcare-associated diarrhoea. Formation of biofilms may protect C. difficile against antibiotics, potentially leading to treatment failure. Furthermore, bacterial spores or vegetative cells may linger in biofilms in the gut causing C. difficile infection recurrence. OBJECTIVES: In this study, we evaluated and compared the efficacy of four antibiotics (fidaxomicin, surotomycin, vancomycin and metronidazole) in penetrating C. difficile biofilms and killing vegetative cells. METHODS: C. difficile biofilms grown initially for 48 or 72 h using the colony biofilm model were then treated with antibiotics at a concentration of 25 × MIC for 24 h. Vegetative cells and spores were enumerated. The effect of treatment on biofilm structure was studied by scanning electron microscopy (SEM). The ability of fidaxomicin and surotomycin to penetrate biofilms was studied using fluorescently tagged antibiotics. RESULTS: Both surotomycin and fidaxomicin were significantly more effective than vancomycin or metronidazole (P < 0.001) at killing vegetative cells in established biofilms. Fidaxomicin was more effective than metronidazole at reducing viable spore counts in biofilms (P < 0.05). Fluorescently labelled surotomycin and fidaxomicin penetrated C. difficile biofilms in < 1 h. After 24 h of treatment, SEM demonstrated that both fidaxomicin and surotomycin disrupted the biofilm structure, while metronidazole had no observable effect. CONCLUSIONS: Fidaxomicin is effective in disrupting C. difficile biofilms, killing vegetative cells and decreasing spore counts.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Clostridioides difficile/drug effects , Lipopeptides/pharmacology , Metronidazole/pharmacology , Peptides, Cyclic/pharmacology , Vancomycin/pharmacology , Biological Transport/physiology , Clostridioides difficile/growth & development , Colony Count, Microbial , Fidaxomicin , Humans , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Spores, Bacterial/drug effects
5.
PLoS One ; 12(1): e0170258, 2017.
Article in English | MEDLINE | ID: mdl-28103289

ABSTRACT

BACKGROUND: Clostridium difficile infection (CDI) is characterized by high rates of recurrence, resulting in substantial health care costs. The aim of this study was to analyze the cost-effectiveness of treatments for the management of second recurrence of community-onset CDI in France. METHODS: We developed a decision-analytic simulation model to compare 5 treatments for the management of second recurrence of community-onset CDI: pulsed-tapered vancomycin, fidaxomicin, fecal microbiota transplantation (FMT) via colonoscopy, FMT via duodenal infusion, and FMT via enema. The model outcome was the incremental cost-effectiveness ratio (ICER), expressed as cost per quality-adjusted life year (QALY) among the 5 treatments. ICERs were interpreted using a willingness-to-pay threshold of €32,000/QALY. Uncertainty was evaluated through deterministic and probabilistic sensitivity analyses. RESULTS: Three strategies were on the efficiency frontier: pulsed-tapered vancomycin, FMT via enema, and FMT via colonoscopy, in order of increasing effectiveness. FMT via duodenal infusion and fidaxomicin were dominated (i.e. less effective and costlier) by FMT via colonoscopy and FMT via enema. FMT via enema compared with pulsed-tapered vancomycin had an ICER of €18,092/QALY. The ICER for FMT via colonoscopy versus FMT via enema was €73,653/QALY. Probabilistic sensitivity analysis with 10,000 Monte Carlo simulations showed that FMT via enema was the most cost-effective strategy in 58% of simulations and FMT via colonoscopy was favored in 19% at a willingness-to-pay threshold of €32,000/QALY. CONCLUSIONS: FMT via enema is the most cost-effective initial strategy for the management of second recurrence of community-onset CDI at a willingness-to-pay threshold of €32,000/QALY.


Subject(s)
Clostridioides difficile , Community-Acquired Infections/economics , Community-Acquired Infections/therapy , Enterocolitis, Pseudomembranous/economics , Enterocolitis, Pseudomembranous/therapy , Aminoglycosides/economics , Aminoglycosides/therapeutic use , Anti-Bacterial Agents/economics , Anti-Bacterial Agents/therapeutic use , Computer Simulation , Cost-Benefit Analysis , Decision Trees , Fecal Microbiota Transplantation/economics , Fecal Microbiota Transplantation/methods , Fidaxomicin , France , Health Care Costs , Humans , Models, Economic , Quality-Adjusted Life Years , Recurrence , Vancomycin/economics , Vancomycin/therapeutic use
6.
Nat Prod Commun ; 11(5): 569-72, 2016 May.
Article in English | MEDLINE | ID: mdl-27319118

ABSTRACT

The co-identity of the antibiotics lipiarmycin A3 obtained from Actinoplanes deccanensis and tiacumicin B obtained from Dactylosporangium aurantiacum was unambiguously demonstrated through a number of experimental means. Spectroscopic analyses performed on both the antibiotics themselves and on their derivatives showed no difference between the two series of compounds. Moreover, unambiguous confirmation of the postulated identity of the two compounds was achieved by chemical degradation of lipiarmycin A3 and isolation of (3S,4R)-pentane-1,3,4-triol triacetate whose relative configuration was assigned by comparison with the authentic erythro and threo pentane-1,3,4-triol triacetates, obtained by chemical synthesis.


Subject(s)
Aminoglycosides/chemistry , Fidaxomicin , Molecular Structure , Terminology as Topic
7.
Anaerobe ; 40: 10-4, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27108094

ABSTRACT

Clostridium difficile is a significant cause of nosocomial-acquired infection that results in severe diarrhea and can lead to mortality. Treatment options for C. difficile infection (CDI) are limited, however, new antibiotics are being developed. Current methods for determining efficacy of experimental antibiotics on C. difficile involve antibiotic killing rates and do not give insight into the drug's pharmacologic effects. Considering this, we hypothesized that by using scanning electron microscopy (SEM) in tandem to drug killing curves, we would be able to determine efficacy and visualize the phenotypic response to drug treatment. To test this hypothesis, supraMIC kill curves were conducted using vancomycin, metronidazole, fidaxomicin, and ridinilazole. Following collection, cells were either plated or imaged using a scanning electron microscope (SEM). Consistent with previous reports, we found that the tested antibiotics had significant bactericidal activity at supraMIC concentrations. By SEM imaging and using a semi-automatic pipeline for image analysis, we were able to determine that vancomycin and to a lesser extent fidaxomicin and ridinilazole significantly affected the cell wall, whereas metronidazole, fidaxomicin, and ridinilazole had significant effects on cell length suggesting a metabolic effect. While the phenotypic response to drug treatment has not been documented previously in this manner, the results observed are consistent with the drug's mechanism of action. These techniques demonstrate the versatility and reliability of imaging and measurements that could be applied to other experimental compounds. We believe the strategies laid out here are vital for characterizing new antibiotics in development for treating CDI.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Wall/drug effects , Clostridioides difficile/drug effects , Optical Imaging/methods , Agar/chemistry , Aminoglycosides/pharmacology , Cell Wall/ultrastructure , Clostridioides difficile/ultrastructure , Culture Media/chemistry , Fidaxomicin , Metronidazole/pharmacology , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Vancomycin/pharmacology
8.
Antimicrob Agents Chemother ; 60(7): 3988-93, 2016 07.
Article in English | MEDLINE | ID: mdl-27090175

ABSTRACT

The use of oral vancomycin or metronidazole for treatment of Clostridium difficile infection (CDI) may promote colonization by health care-associated pathogens due to disruption of the intestinal microbiota. Because the macrocyclic antibiotic fidaxomicin causes less alteration of the intestinal microbiota than vancomycin, we hypothesized that it would not lead to a loss of colonization resistance to vancomycin-resistant enterococci (VRE) and extended-spectrum-ß-lactamase-producing Klebsiella pneumoniae (ESBL-Kp). Mice (8 per group) received orogastric saline, vancomycin, or fidaxomicin daily for 5 days at doses resulting in stool concentrations in mice similar to those measured in humans. The mice were challenged with 10(5) CFU of orogastric VRE or ESBL-Kp on day 2 of treatment and concentrations of the pathogens in stool were monitored. The impact of drug exposure on the microbiome was measured by cultures, real-time PCR for selected anaerobic bacteria, and deep sequencing. In comparison to saline controls, oral vancomycin promoted establishment of high-density colonization by VRE and ESBL-Kp in stool (8 to 10 log10 CFU/g; P < 0.001), whereas fidaxomicin did not (<4 log10 CFU; P > 0.5). Vancomycin treatment resulted in significant reductions in enterococci, Bacteroides spp., and Clostridium leptum, whereas the population of aerobic and facultative Gram-negative bacilli increased; deep-sequencing analysis demonstrated suppression of Firmicutes and expansion of Proteobacteria during vancomycin treatment. Fidaxomicin did not cause significant alteration of the microbiota. In summary, in contrast to vancomycin, fidaxomicin treatment caused minimal disruption of the intestinal microbiota and did not render the microbiota susceptible to VRE and ESBL-Kp colonization.


Subject(s)
Aminoglycosides/therapeutic use , Anti-Bacterial Agents/therapeutic use , Intestines/microbiology , Klebsiella pneumoniae/drug effects , Vancomycin-Resistant Enterococci/drug effects , Vancomycin/therapeutic use , Aminoglycosides/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bacteroides Infections/drug therapy , Bacteroides Infections/microbiology , Clostridium/drug effects , Clostridium/pathogenicity , Feces/microbiology , Female , Fidaxomicin , Firmicutes/drug effects , Firmicutes/pathogenicity , Klebsiella pneumoniae/pathogenicity , Mice , Microbial Sensitivity Tests , Real-Time Polymerase Chain Reaction , Vancomycin/pharmacology , Vancomycin-Resistant Enterococci/pathogenicity , beta-Lactamases/genetics , beta-Lactamases/metabolism
9.
Cold Spring Harb Perspect Med ; 6(2): a025445, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26834162

ABSTRACT

Clostridium difficile is the most common cause of antibacterial-associated diarrhea. Clear clinical presentation and rapid diagnostics enable targeted therapy for C. difficile infection (CDI) to start quickly. CDI treatment includes metronidazole and vancomycin (VAN). Despite decades of use for CDI, no clinically meaningful resistance to either agent has emerged. Fidaxomicin (FDX), an RNA polymerase inhibitor, is also approved to treat CDI. Mutants with reduced susceptibility to FDX have been selected in vitro by single and multistep methods. Strains with elevated FDX minimum inhibitory concentrations (MICs) were also identified from FDX-treated patients in clinical trials. LFF571 is an exploratory agent that inhibits EF-Tu. In a proof-of-concept study, LFF571 was safe and effective for treating CDI. Spontaneous mutants with reduced susceptibility to LFF571 were selected in vitro in a single step, but not via serial passage. Although there are several agents in development for treatment of CDI, this review summarizes the frequencies and mechanisms of C. difficile mutants displaying reduced susceptibility to FDX or LFF71.


Subject(s)
Aminoglycosides/therapeutic use , Anti-Bacterial Agents/therapeutic use , Clostridium Infections/drug therapy , Thiazoles/therapeutic use , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Clinical Trials, Phase III as Topic , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Drug Resistance, Bacterial/genetics , Fidaxomicin , Genotype , Humans , Microbial Sensitivity Tests , Mutation , Thiazoles/chemistry , Thiazoles/pharmacology
10.
Expert Rev Anti Infect Ther ; 14(2): 193-206, 2016.
Article in English | MEDLINE | ID: mdl-26641310

ABSTRACT

Diarrhoea is one of the most commonly occurring diseases. This article presents a review of the current state of the treatment of acute infectious diarrhoea, as well as of the most important pathogens. The general principles of the therapy of diarrhoea are exemplified, followed by a description of the targeted antimicrobial therapy of the most important bacterial gastrointestinal infections, including salmonellosis, shigellosis and Campylobacter infections, as well as infections with pathogenic Escherichia coli strains, yersiniosis and cholera. Diarrhoea caused by toxigenic Clostridium difficile strains has increased in incidence and in severity. These infections will therefore be described in detail, including important new aspects of treatment. Symptomatic therapy is still the most important component of the treatment of infectious diarrhoea. However, empirical antibiotic therapy should be considered for severely ill patients with a high frequency of stools, fever, bloody diarrhoea, underlying immune deficiency, advanced age or significant comorbidities. Increasing resistance, in particular against fluoroquinolones, must be taken into consideration. Therapy with motility inhibitors is not recommended for Shiga toxin-producing Escherichia coli (STEC) infections, Clostridium difficile infections (CDI), and severe colitis. The macrocyclic antibiotic fidaxomicin can reduce the rate of recurrent disease in CDI. Furthermore, evidence for the benefits of faecal microbiota transplantation as a treatment option for multiple recurrences of CDI is increasing. In conclusion, the treatment of acute diarrhoea is still primarily supportive. General empirical antibiotic therapy for acute diarrhoea is not evidence-based.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Diarrhea/drug therapy , Gastroenteritis/drug therapy , Acute Disease , Aminoglycosides/therapeutic use , Azithromycin/therapeutic use , Bacterial Infections/diagnosis , Campylobacter Infections/diagnosis , Campylobacter Infections/drug therapy , Cholera/diagnosis , Cholera/drug therapy , Ciprofloxacin/therapeutic use , Dysbiosis/chemically induced , Dysentery, Bacillary/diagnosis , Dysentery, Bacillary/drug therapy , Enterocolitis, Pseudomembranous/diagnosis , Enterocolitis, Pseudomembranous/drug therapy , Escherichia coli Infections/diagnosis , Escherichia coli Infections/drug therapy , Fidaxomicin , Humans , Rifamycins/therapeutic use , Rifaximin , Salmonella Infections/diagnosis , Salmonella Infections/drug therapy , Shiga-Toxigenic Escherichia coli , Yersinia Infections/diagnosis , Yersinia Infections/drug therapy
12.
Nat Prod Commun ; 9(2): 237-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24689300

ABSTRACT

The co-identity of the antibiotics lipiarmycin A3 obtained from Actinoplanes deccanensis and tiacumicin B obtained from Dactylosporangium aurantiacum was unambiguously demonstrated through a number of experimental means. Spectroscopic analyses performed on both the antibiotics themselves and on their derivatives showed no difference between the two series of compounds. Moreover, unambiguous confirmation of the postulated identity of the two compounds was achieved by chemical degradation of lipiarmycin A3 and isolation of (3S,4R)-pentane-1,3,4-triol triacetate whose relative configuration was assigned by comparison with the authentic erythro and threo pentane-1,3,4-triol triacetates, obtained by chemical synthesis.


Subject(s)
Aminoglycosides/isolation & purification , Anti-Bacterial Agents/isolation & purification , Aminoglycosides/chemistry , Fidaxomicin , Molecular Conformation
13.
Antimicrob Agents Chemother ; 58(2): 901-8, 2014.
Article in English | MEDLINE | ID: mdl-24277035

ABSTRACT

Cadazolid is a new oxazolidinone-type antibiotic currently in clinical development for the treatment of Clostridium difficile-associated diarrhea. Here, we report investigations on the mode of action and the propensity for spontaneous resistance development in C. difficile strains. Macromolecular labeling experiments indicated that cadazolid acts as a potent inhibitor of protein synthesis, while inhibition of DNA synthesis was also observed, albeit only at substantially higher concentrations of the drug. Strong inhibition of protein synthesis was also obtained in strains resistant to linezolid, in agreement with low MICs against such strains. Inhibition of protein synthesis was confirmed in coupled transcription/translation assays using extracts from different C. difficile strains, including strains resistant to linezolid, while inhibitory effects in DNA topoisomerase assays were weak or not detectable under the assay conditions. Spontaneous resistance frequencies of cadazolid were low in all strains tested (generally <10(-10) at 2× to 4× the MIC), and in multiple-passage experiments (up to 13 passages) MICs did not significantly increase. Furthermore, no cross-resistance was observed, as cadazolid retained potent activity against strains resistant or nonsusceptible to linezolid, fluoroquinolones, and the new antibiotic fidaxomicin. In conclusion, the data presented here indicate that cadazolid acts primarily by inhibition of protein synthesis, with weak inhibition of DNA synthesis as a potential second mode of action, and suggest a low potential for spontaneous resistance development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Drug Resistance, Bacterial/genetics , Protein Biosynthesis/drug effects , Acetamides/pharmacology , Aminoglycosides/pharmacology , Clostridioides difficile/genetics , Clostridioides difficile/growth & development , Clostridioides difficile/metabolism , DNA Gyrase/genetics , DNA Gyrase/metabolism , Drug Resistance, Bacterial/drug effects , Fidaxomicin , Fluoroquinolones/pharmacology , Linezolid , Microbial Sensitivity Tests , Oxazolidinones/pharmacology , Protein Biosynthesis/genetics , RNA/antagonists & inhibitors , RNA/biosynthesis , Recombinant Proteins , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism , Transcription, Genetic/drug effects , Vancomycin/pharmacology
14.
Drugs ; 73(15): 1733-47, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24136090

ABSTRACT

Oral fidaxomicin (Dificid(®); Dificlir(®)) is a first-in-class macrocyclic antibacterial that is approved in several countries for the treatment of adult patients with Clostridium difficile-associated diarrhoea. Fidaxomicin 200 mg twice daily for 10 days was an effective and generally well tolerated treatment in adult patients with a first episode or first recurrence of C. difficile infection. In two multinational phase III trials, fidaxomicin treatment was noninferior to vancomycin treatment with regard to clinical cure rates and was associated with statistically significantly lower C. difficile infection recurrence rates and statistically significantly higher global cure rates than vancomycin. The drug has a favourable pharmacological profile, including having a narrow spectrum of activity that targets relevant pathogens, minimal impact on normal faecal microflora, a convenient treatment regimen and attainment of very high faecal concentrations. Albeit further clinical experience is required to fully define the position of fidaxomicin, it is a valuable emerging option for the treatment of first episode and recurrent episodes of C. difficile-associated diarrhoea.


Subject(s)
Aminoglycosides/therapeutic use , Anti-Bacterial Agents/therapeutic use , Clostridioides difficile/drug effects , Enterocolitis, Pseudomembranous/drug therapy , Aminoglycosides/administration & dosage , Aminoglycosides/adverse effects , Aminoglycosides/pharmacokinetics , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacokinetics , Clinical Trials, Phase III as Topic , Clostridioides difficile/isolation & purification , Drug Resistance, Multiple, Bacterial , Enterocolitis, Pseudomembranous/microbiology , Fidaxomicin , Humans , Microbial Sensitivity Tests , Treatment Outcome
15.
Clin Gastroenterol Hepatol ; 11(10): 1216-23; quiz e73, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23542332

ABSTRACT

Clostridium difficile infection (CDI) is increasing in frequency and severity in and out of the hospital, with a high probability of recurrence after treatment. The recent literature on CDI was reviewed using PubMed to include recent publications dealing with diagnosis and therapy. Real-time polymerase chain reaction is a sensitive and useful diagnostic test for CDI but there are growing concerns of false-positive test results if the rate of CDI is low in the patient population providing samples and/or if the population being studied commonly includes people with C difficile colonization. Recommended therapy of CDI includes oral metronidazole for milder cases of CDI and oral vancomycin or fidaxomicin for more severe cases, each given for 10 days. Colectomy is being performed more frequently in patients with fulminant CDI. For treatment of first recurrences the drug used in the first bout can be used again and for second recurrences longer courses of vancomycin often are given in a tapered dose or intermittently to allow gut flora reconstitution, or other treatments including fidaxomicin may be used. Bacteriotherapy with fecal transplantation is playing an increasing role in therapy of recurrent cases. Metagenomic studies of patients with CDI during successful therapy are needed to determine how best to protect the flora from assaults from antibacterial drugs and to develop optimal therapeutic approaches. Immunotherapy and immunoprophylaxis offer opportunities to prevent CDI, to speed up recovery from CDI, and to eliminate recurrent infection. Humanized monoclonal antitoxin antibodies and active immunization with vaccines against C difficile or its toxins are both in development and appear to be of potential value.


Subject(s)
Clostridioides difficile/isolation & purification , Clostridium Infections/diagnosis , Clostridium Infections/drug therapy , Administration, Oral , Aminoglycosides/therapeutic use , Anti-Infective Agents/therapeutic use , Biological Therapy/methods , Clostridium Infections/microbiology , Colectomy , False Positive Reactions , Fidaxomicin , Humans , Immunotherapy/methods , Metronidazole/therapeutic use , Molecular Diagnostic Techniques/methods , Vancomycin/therapeutic use
16.
J Antimicrob Chemother ; 68(6): 1305-11, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23420839

ABSTRACT

OBJECTIVES: In November 2008, a study was performed with support from the European Centre for Disease Prevention and Control (ECDC) to obtain an overview of Clostridium difficile infections (CDIs) in European hospitals. A collection of 398 C. difficile isolates obtained from this hospital-based survey was utilized to identify antimicrobial susceptibility patterns of common C. difficile PCR ribotypes across Europe. METHODS: The MICs of three approved therapeutic agents (vancomycin, metronidazole and fidaxomicin) and LFF571 (a novel semi-synthetic thiopeptide antibiotic) were determined by the agar dilution method. RESULTS: MICs of fidaxomicin and LFF571 were in general 2-4-fold lower than those of vancomycin and metronidazole. Isolates belonging to clade 2, including the hypervirulent ribotype 027, had one-dilution higher MIC50 and MIC90 values for fidaxomicin and metronidazole, whereas similar MIC values were observed for vancomycin and LFF571. Isolates belonging to C. difficile PCR ribotype 001 were more susceptible to fidaxomicin than other frequently found PCR ribotypes 014/020 and 078. Six isolates from three different countries had a metronidazole MIC of 2 mg/L. Four of the six isolates were characterized as PCR ribotype 001. CONCLUSIONS: There was no evidence of in vitro resistance of C. difficile to any of the four agents tested. However, the results suggest type-specific differences in susceptibility for the treatment agents we investigated. Continuous surveillance of C. difficile isolates in Europe is needed to determine the possible clinical implications of ribotype-specific changes in susceptibility to therapeutic agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Thiazoles/pharmacology , Aminoglycosides/pharmacology , Aminoglycosides/therapeutic use , Anti-Bacterial Agents/therapeutic use , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Cross Infection/drug therapy , Cross Infection/microbiology , Enterocolitis, Pseudomembranous/drug therapy , Enterocolitis, Pseudomembranous/microbiology , Europe , Fidaxomicin , Humans , Metronidazole/therapeutic use , Microbial Sensitivity Tests , Multilocus Sequence Typing , Polymerase Chain Reaction , Ribotyping , Thiazoles/therapeutic use , Treatment Outcome , Vancomycin/pharmacology , Vancomycin/therapeutic use
17.
Curr Protoc Microbiol ; 30: 9A.3.1-9A.3.9, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24510892

ABSTRACT

Metronidazole and vancomycin remain the front-line therapies for most Clostridium difficile infections (CDI). However, recurrent CDI occurs in ∼ 25% of patients, causing significant morbidity and mortality and healthcare costs. For this population, traditional antibiotic therapies fail and new treatment options are greatly needed. The US Food and Drug Administration recently approved fidaxomicin for CDI treatment. This narrow-spectrum antibiotic preserves the normal gut microbiota and shows promise as a treatment for severe and recurrent CDI. Monoclonal antibodies and vaccines directed against toxin are currently in clinical trials and represent alternative, non-antibiotic therapies. Less traditional therapeutic interventions include bacteriotherapy with non-toxigenic C. difficile and fecal transplant. This commentary will provide an overview of current and forthcoming CDI therapies.


Subject(s)
Clostridioides difficile/isolation & purification , Clostridium Infections/microbiology , Clostridium Infections/therapy , Diarrhea/drug therapy , Diarrhea/microbiology , Aminoglycosides/therapeutic use , Anti-Infective Agents/therapeutic use , Antibodies, Bacterial/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antitoxins/therapeutic use , Bacterial Vaccines/therapeutic use , Biological Therapy/methods , Drug Therapy/methods , Fidaxomicin , Humans , Immunotherapy/methods , Metronidazole/therapeutic use , Vancomycin/therapeutic use
18.
Clin Infect Dis ; 55 Suppl 2: S110-5, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22752858

ABSTRACT

Fidaxomicin is a novel macrocyclic antibiotic recently approved by the US Food and Drug Administration for the treatment of Clostridium difficile-associated diarrhea in adults. We reviewed safety data from nonclinical studies and clinical trials (phases 1, 2A, and 3) with fidaxomicin. In nonclinical studies, fidaxomicin was administered orally at approximately 1 g/kg/d to dogs for up to 3 months with no significant target-organ toxicities observed. A total of 728 adults have received oral fidaxomicin in clinical trials to date: 116 healthy volunteers and 612 patients with C. difficile infection. In phase 3 clinical trials, fidaxomicin was well tolerated, with a safety profile comparable with oral vancomycin. There were no differences in the incidence of death or serious adverse events between the 2 drugs. Fidaxomicin appears to be well tolerated. Continued monitoring of adverse events in the postmarketing setting will provide additional information about the full safety profile of fidaxomicin.


Subject(s)
Aminoglycosides/therapeutic use , Clostridioides difficile/pathogenicity , Clostridium Infections/drug therapy , Vancomycin/therapeutic use , Aminoglycosides/administration & dosage , Aminoglycosides/adverse effects , Animals , Anti-Bacterial Agents/therapeutic use , Clinical Trials as Topic , Clostridium Infections/microbiology , Diarrhea/microbiology , Dogs , Drug Evaluation, Preclinical , Drug-Related Side Effects and Adverse Reactions , Fidaxomicin , Humans , Treatment Outcome
19.
Clin Infect Dis ; 55 Suppl 2: S143-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22752863

ABSTRACT

Fidaxomicin is bactericidal against Clostridium difficile. The combined results of 8 in vitro studies of 1323 C. difficile isolates showed the minimum inhibitory concentration (MIC) range of fidaxomicin to be ≤ 0.001-1 µg/mL, with a maximum MIC for inhibition of 90% of organisms (MIC(90)) of 0.5 µg/mL. Isolates from 2 phase III clinical trials demonstrated that fidaxomicin MICs of baseline isolates did not predict clinical cure, failure, or recurrence of C. difficile infections. No resistance to fidaxomicin developed during treatment in either study, although a single strain recovered from a cured patient had an elevated MIC of 16 µg/mL at the time of recurrence. For 135 strains, OP-1118, a major metabolite, had an MIC for inhibition of 50% of organisms of 4 µg/mL and an MIC(90) of 8 µg/mL. Changes in inoculum size (10(2)-10(5) colony-forming units/spot) or cation concentrations of calcium or magnesium appeared to have no effect on fidaxomicin MICs. Fidaxomicin has little or no activity against gram-negative aerobes and anaerobes or yeast.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clinical Trials as Topic , Clostridioides difficile/isolation & purification , Clostridium Infections/microbiology , Drug Evaluation, Preclinical , Drug Resistance, Bacterial , Feces/microbiology , Fidaxomicin , Humans , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Recurrence , Vancomycin/pharmacology
20.
Am J Health Syst Pharm ; 69(11): 933-43, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22610025

ABSTRACT

PURPOSE: The pharmacology, clinical efficacy, safety, dosage and administration, and place in therapy of fidaxomicin for the treatment of Clostridium difficile infection (CDI) are reviewed. SUMMARY: Fidaxomicin, a macrocyclic antibiotic, has a narrow spectrum of activity against gram-positive anaerobes and is bactericidal against C. difficile. It has no activity against gram-negative bacteria. Fidaxomicin has minimal activity against Bacteroides species, which may be advantageous in maintaining colonization resistance and protecting the gastrointestinal tract from colonization by C. difficile. The minimum inhibitory concentration for 90% of organisms for fidaxomicin against C. difficile ranged from 0.0078 to 2 µg/mL in in vitro studies. After oral administration, fecal concentrations are detected and are directly proportional to the dose administered. Fidaxomicin resistance in vivo has not been reported. In clinical trials, fidaxomicin has been shown to be noninferior to vancomycin in the management of mild-to-moderately severe CDI. The adverse-effect profile of fidaxomicin is comparable to that of vancomycin. The recommended dosage for treatment of CDI is fidaxomicin 200 mg orally twice daily for 10 days. Fidaxomicin should be considered for patients who previously received treatment with metronidazole or vancomycin for CDI and who are diagnosed with recurrent CDI in which a non-NAP1/BI/027 strain is isolated. At institutions where strain typing is not available, fidaxomicin may be considered in patients with recurrent CDI who have not responded to treatment with the regimen used for the first episode of CDI. CONCLUSION: Fidaxomicin is a well-tolerated agent for the treatment of CDI and has been shown to be noninferior to vancomycin in the management of mild-to-moderately severe CDI.


Subject(s)
Aminoglycosides/therapeutic use , Anti-Bacterial Agents/therapeutic use , Clostridium Infections/drug therapy , Aminoglycosides/administration & dosage , Aminoglycosides/adverse effects , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Clostridioides difficile/drug effects , Clostridium Infections/microbiology , Fidaxomicin , Humans , Microbial Sensitivity Tests , Severity of Illness Index , Vancomycin/adverse effects , Vancomycin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL