Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Publication year range
1.
J Antimicrob Chemother ; 75(11): 3120-3125, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32747931

ABSTRACT

OBJECTIVES: To investigate the molecular epidemiology and antimicrobial susceptibility of Clostridioides difficile isolates from patients with C. difficile infection (CDI) from two Phase 3 clinical trials of surotomycin. METHODS: In both trials [Protocol MK-4261-005 (NCT01597505) conducted across Europe, North America and Israel; and Protocol MK-4261-006 (NCT01598311) conducted across North America, Asia-Pacific and South America], patients with CDI were randomized (1:1) to receive oral surotomycin (250 mg twice daily) or oral vancomycin (125 mg four times per day) for 10 days. Stool samples were collected at baseline and C. difficile isolates were characterized by restriction endonuclease analysis (REA) and PCR ribotyping. Susceptibility testing was performed by agar dilution, according to CLSI recommendations. RESULTS: In total, 1147 patients were included in the microbiological modified ITT population. Of 992 recovered isolates, 922 (92.9%) were typed. There was a high association between REA groups and their corresponding predominant PCR ribotype (RT) for BI, DH, G and CF strains. REA group A showed more diverse PCR RTs. Overall, the most common strain was BI/RT027 (20.3%) followed by Y/RT014/020 (15.0%) and DH/RT106 (7.2%). The BI/RT027 strain was particularly prevalent in Europe (29.9%) and Canada (23.6%), with lower prevalence in the USA (16.8%) and Australia/New Zealand (3.4%). Resistance was most prevalent in the BI/RT027 strain, particularly to metronidazole, vancomycin and moxifloxacin. CONCLUSIONS: A wide variation in C. difficile strains, both within and across different geographical regions, was documented by both REA and ribotyping, which showed overall good correlation.


Subject(s)
Anti-Infective Agents , Clostridioides difficile , Clostridium Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Asia , Canada , Clostridioides , Clostridioides difficile/genetics , Clostridium Infections/drug therapy , Clostridium Infections/epidemiology , DNA Restriction Enzymes , Europe , Humans , Israel , Lipopeptides , Microbial Sensitivity Tests , North America , Peptides, Cyclic , Polymerase Chain Reaction , Prohibitins , Ribotyping , South America
2.
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
3.
Anaerobe ; 53: 38-42, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29886050

ABSTRACT

The prevalence of C. difficile infection (CDI) and severe CDI are influenced by the prevalence of specific C. difficile strains, which are themselves influenced by antimicrobial susceptibility determinants as well as antimicrobial usage patterns. Restriction endonuclease analysis (REA) typing and antimicrobial susceptibility testing were used to characterize 1808 C. difficile isolates obtained from patients enrolled in four multicenter, multi-country, randomized CDI treatment trials conducted between 2006 and 2009 and between 2012 and 2015. By 2015, the epidemic REA group BI strain (RT027) had decreased in prevalence in North America (US: 43%-18%, Canada: 39%-24%, P < 0.001), but rates of moxifloxacin resistance remained high. In contrast, REA group Y (RT014/020) and DH (RT106) strains, both of which had low rates of moxifloxacin resistance, increased in prevalence (Y strain - US: 6%-17%, Canada: 11%-23%, P < 0.001; DH strain - US: 1%-11%, Canada: 0%-8%, P < 0.0001). In Europe, the BI strain (RT027) was highly prevalent in Eastern European countries in 2015, but was unchanged in other parts of Europe. As in North America, the Y strain (RT014/020) was prevalent in both time periods, but the DH strain was rarely identified. Continued international molecular surveillance of C. difficile will be important to track prevalence of known epidemic strains and detect emergence of new strains of potential epidemiologic significance.


Subject(s)
Clostridioides difficile/classification , Clostridioides difficile/genetics , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Genotype , Canada/epidemiology , Clostridioides difficile/isolation & purification , Drug Resistance, Bacterial , Europe/epidemiology , Humans , Microbial Sensitivity Tests , Molecular Epidemiology , Molecular Typing , Prevalence , Prohibitins , Randomized Controlled Trials as Topic , United States/epidemiology
4.
J Antimicrob Chemother ; 72(12): 3453-3461, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28927227

ABSTRACT

OBJECTIVES: The effects of surotomycin (CB-183,315, MK-4261), a bactericidal cyclic lipopeptide, and vancomycin, the current standard-of-care for Clostridium difficile infection (CDI), on intestinal pathogens and microbiota were evaluated parallel to a Phase 2 randomized, double-blind clinical trial. METHODS: The single-centre cohort included 26 patients receiving surotomycin [125 or 250 mg twice daily (n = 9 each)] or oral vancomycin [125 mg four times daily (n = 8)] for 10 days. Faecal samples were collected at days 0-42 to quantify both C. difficile by conventional culture and the major components of the microbiome by quantitative PCR. RESULTS: Surotomycin 250 mg twice daily or vancomycin 125 mg four times daily reduced faecal C. difficile counts from ∼105-107 log10 cfu/g at baseline to ≤ 102 cfu/g by days 4-10 of treatment. Day 10 counts of C. difficile in 3/9 patients receiving surotomycin 125 mg twice daily remained detectable, including one patient who failed to achieve clinical cure. Bacteroidetes and Prevotella mean counts increased 0.7 log10 or remained unchanged with surotomycin 125 and 250 mg twice daily, respectively, whereas vancomycin reduced counts by 2.5-3.2 log10 (P < 0.02). Vancomycin reduced Firmicutes counts by 2.5-2.8 log10; surotomycin moderately suppressed these microbes in a dose-dependent manner. CONCLUSIONS: In this Phase 2 trial substudy, compared with vancomycin 125 mg four times daily, surotomycin 250 mg twice daily is as active in vivo against C. difficile, but was more sparing of microbiota. Surotomycin is no longer in development due to failed Phase 3 efficacy results.


Subject(s)
Anti-Bacterial Agents/adverse effects , Bacteria/isolation & purification , Clostridium Infections/drug therapy , Dysbiosis/chemically induced , Gastrointestinal Microbiome/drug effects , Lipopeptides/adverse effects , Peptides, Cyclic/adverse effects , Vancomycin/adverse effects , Administration, Oral , Adolescent , Adult , Aged , Aged, 80 and over , Anti-Bacterial Agents/administration & dosage , Bacteria/classification , Bacterial Load , Bacteriological Techniques , Double-Blind Method , Feces/microbiology , Female , Humans , Lipopeptides/administration & dosage , Male , Metagenomics , Middle Aged , Peptides, Cyclic/administration & dosage , Real-Time Polymerase Chain Reaction , Time Factors , Vancomycin/administration & dosage , Young Adult
5.
Anaerobe ; 44: 73-77, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28188880

ABSTRACT

Due to increasing antibiotic resistance among anaerobic bacteria, routine antimicrobial susceptibility testing is recommended by the Clinical and Laboratory Standards Institute (CLSI). This study compared the minimum inhibitory concentrations (MICs) from 920 Clostridium difficile isolates tested against seven antimicrobial agents using the two current CLSI reference methodologies, agar dilution method, vs broth microdilution method. A subset of isolate testing was performed independently by two laboratories to evaluate reproducibility. A negative bias was noted for MICs generated from broth microdilution compared to agar dilution and the reproducibility was variable and drug dependent. Therefore, broth microdilution is not recommended as an alternative to agar dilution for C. difficile antimicrobial susceptibility testing.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Culture Media/chemistry , Microbial Sensitivity Tests/methods , Clostridioides difficile/isolation & purification , Humans , Reproducibility of Results
6.
Antimicrob Agents Chemother ; 60(6): 3519-23, 2016 06.
Article in English | MEDLINE | ID: mdl-27021314

ABSTRACT

Surotomycin is a cyclic lipopeptide in development for Clostridium difficile-associated diarrhea. This study aimed to assess the impact of surotomycin exposure on C. difficile toxin A and B concentrations and the associated changes in immune response in comparison to vancomycin and metronidazole. Time-kill curve assays were performed using strain R20291 (BI/NAP1/027) at supra-MICs (4× and 40×) and sub-MICs (0.5×) of surotomycin and comparators. Following treatment, CFU counts, toxin A and B concentrations, and cellular morphological changes using scanning electron microscopy were examined. Inflammatory response was determined by measuring interleukin-8 (IL-8) concentrations from polarized Caco-2 cells exposed to antibiotic-treated C. difficile growth media. Supra-MICs (4× and 40×) of surotomycin resulted in a reduction of vegetative cells over 72 h (4-log difference, P < 0.01) compared to controls. These results correlated with decreases of 77% and 68% in toxin A and B production at 48 h, respectively (P < 0.005, each), which resulted in a significant reduction in IL-8 concentration compared to controls. Similar results were observed with comparator antibiotics. Bacterial cell morphology showed that the cell wall was broken apart by surotomycin treatment at supra-MICs while sub-MIC studies showed a "deflated" phenotype plus a rippling effect. These results suggest that surotomycin has potent killing effects on C. difficile that results in reduced toxin production and attenuates the immune response similar to comparator antibiotics. The morphological data also confirm observations that surotomycin is a membrane-active antibiotic.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/biosynthesis , Bacterial Toxins/biosynthesis , Clostridioides difficile/drug effects , Clostridioides difficile/immunology , Enterotoxins/biosynthesis , Lipopeptides/pharmacology , Peptides, Cyclic/pharmacology , Caco-2 Cells , Cell Line, Tumor , Clostridioides difficile/metabolism , Humans , Interleukin-8/immunology , Metronidazole/pharmacology , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Vancomycin/pharmacology
7.
Antimicrob Agents Chemother ; 60(4): 2069-74, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26787687

ABSTRACT

Clostridium difficile-associated diarrhea has been associated with disruption of the normal intestinal microbiota, particularly theBacteroides fragilisgroup andPrevotellaspecies. Surotomycin is a bactericidal cyclic lipopeptide in development for treatment ofClostridium difficile-associated diarrhea that has selective and potent activity againstC. difficileand other Gram-positive bacteria and a minimal impact on intestinal Gram-negative organisms. The impacts of ascending doses of surotomycin on major organism groups in the gut microbiota of healthy volunteers were evaluated during a randomized, double-blind, placebo-controlled, multiple-dose phase 1 study. Thirty volunteers were randomized into 3 cohorts, using a 4:1 ratio, to receive 250 mg, 500 mg, or 1,000 mg of surotomycin, or placebo, twice daily for 14 days. Stool samples collected at baseline (days 0 and 1) and at the end of treatment (days 13 to 15) were cultured quantitatively. TheB. fragilisgroup, theBacteroides/Prevotellagroup, andEnterobacteriaceaewere also quantified by quantitative real-time PCR. Baseline and end-of-treatment stool samples showed 1- to 2-log10CFU/g reductions in total bacterial counts for most volunteers. Various decreases in clostridial,Lactobacillus-Bifidobacteriumgroup, and enterococcus-streptococcus group counts occurred while patients were receiving surotomycin, whereas the enterobacteria and theB. fragilisgroup persisted at the end of treatment. There was no change in enterococcus MICs of surotomycin, nor was vancomycin-resistantEnterococcusdetected after exposure. Surotomycin at doses of up to 1,000 mg twice daily had only modest disruptive effects on the gut microbiota. The potential sparing of the gut microbiota by surotomycin may decrease the risk of disease recurrence.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Lipopeptides/pharmacology , Peptides, Cyclic/pharmacology , Adult , Bacteroides fragilis/drug effects , Bacteroides fragilis/physiology , Double-Blind Method , Enterobacteriaceae/drug effects , Enterobacteriaceae/physiology , Enterococcus/drug effects , Enterococcus/physiology , Feces/microbiology , Female , Gastrointestinal Microbiome/physiology , Healthy Volunteers , Humans , Male , Microbial Sensitivity Tests , Middle Aged , Prevotella/drug effects , Prevotella/physiology
8.
Antimicrob Agents Chemother ; 60(6): 3333-9, 2016 06.
Article in English | MEDLINE | ID: mdl-26976870

ABSTRACT

Surotomycin (formerly called CB-183,315) is a novel, orally administered cyclic lipopeptide antibacterial in development for the treatment of Clostridium difficile infection (CDI) that has potent activity against vancomycin-resistant enterococci (VRE) but limited activity against Gram-negative bacilli, including Bacteroides spp. We used a mouse model to investigate the impact of surotomycin exposure on the microbiome, and to test the consequences of the disruption on colonization by vancomycin-resistant enterococci (VRE) and extended-spectrum ß-lactamase-producing Klebsiella pneumoniae (ESBL-KP), in comparison with the effects of oral vancomycin and metronidazole. Mice (8 per group) received saline, vancomycin, metronidazole, or surotomycin through an orogastric tube daily for 5 days and were challenged with 10(5) CFU of VRE or ESBL-KP administered through an orogastric tube on day 2 of treatment. The concentrations of the pathogens in stool were determined during and after treatment by plating on selective media. A second experiment was conducted to determine if the antibiotics would inhibit established VRE colonization. In comparison to controls, oral vancomycin promoted VRE and ESBL-KP overgrowth in stool (8 log10 to 10 log10 CFU/g; P < 0.001), whereas metronidazole did not (<4 log10 CFU/g; P > 0.5). Surotomycin promoted ESBL-KP overgrowth (>8 log10 CFU/g; P, <0.001 for comparison with saline controls) but not VRE overgrowth. Surotomycin suppressed preexisting VRE colonization, whereas metronidazole and vancomycin did not. These results suggest that treatment of CDI with surotomycin could reduce levels of VRE acquisition and overgrowth from those with agents such as vancomycin and metronidazole. However, surotomycin and vancomycin may promote colonization by antibiotic-resistant Gram-negative bacilli.


Subject(s)
Anti-Bacterial Agents/pharmacology , Klebsiella pneumoniae/drug effects , Lipopeptides/pharmacology , Peptides, Cyclic/pharmacology , Vancomycin-Resistant Enterococci/drug effects , Animals , Female , Gram-Negative Bacteria/drug effects , Metronidazole/pharmacology , Mice , Vancomycin/pharmacology , Vancomycin Resistance
9.
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
10.
J Clin Microbiol ; 54(3): 749-53, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26677246

ABSTRACT

Test parameter variations were evaluated for their effects on surotomycin MICs. Calcium concentration was the only variable that influenced MICs; therefore, 50 µg/ml (standard for lipopeptide testing) is recommended. Quality control ranges for Clostridium difficile (0.12 to 1 µg/ml) and Eggerthella lenta (broth, 1 to 4 µg/ml; agar, 1 to 8 µg/ml) were approved by the Clinical and Laboratory Standards Institute based on these data.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Lipopeptides/pharmacology , Microbial Sensitivity Tests/methods , Microbial Sensitivity Tests/standards , Peptides, Cyclic/pharmacology , Humans , Quality Control , Reproducibility of Results , Sensitivity and Specificity
11.
J Antimicrob Chemother ; 71(10): 2964-71, 2016 10.
Article in English | MEDLINE | ID: mdl-27432604

ABSTRACT

OBJECTIVES: Clostridium difficile infection (CDI) is a major public health concern. Treatment with commonly prescribed antibiotics is associated with high rates of recurrence after initial cure. Here, we present the efficacy and safety of surotomycin, an orally administered, minimally absorbed, selective bactericidal cyclic lipopeptide, compared with vancomycin, in patients with CDI. METHODS: In this Phase 2, randomized, controlled, double-blind, non-inferiority, multicentre trial, participants received surotomycin 125 mg twice daily, surotomycin 250 mg twice daily or vancomycin 125 mg four times daily for 10 days. The primary efficacy outcome was clinical response at end of treatment. The registration number of the study on clinicaltrials.gov is NCT01085591. RESULTS: Clinical cure rates were similar among treatment groups (92.4% for surotomycin 125 mg twice daily, 86.6% for surotomycin 250 mg twice daily and 89.4% for vancomycin). Recurrence rates were 27.9% for surotomycin 125 mg twice daily, 17.2% for surotomycin 250 mg twice daily and 35.6% for vancomycin. The lower recurrence rate with surotomycin 250 mg twice daily versus vancomycin was statistically significant (P = 0.035). Recurrence rates were statistically similar between the surotomycin dose groups (P = 0.193). Rates of sustained clinical response at end of study were 66.7% for surotomycin 125 mg twice daily, 70.1% for surotomycin 250 mg twice daily and 56.1% for vancomycin. Incidence of adverse events was similar among treatment arms. CONCLUSIONS: Recurrence rates of CDI were lower with surotomycin with higher sustained clinical response rates compared with vancomycin, both of which may offer potential clinical benefits.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Lipopeptides/therapeutic use , Peptides, Cyclic/therapeutic use , Vancomycin/therapeutic use , Administration, Oral , Adult , Aged , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Clostridium Infections/microbiology , Double-Blind Method , Female , Humans , Lipopeptides/administration & dosage , Lipopeptides/adverse effects , Male , Middle Aged , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/adverse effects , Recurrence , Treatment Outcome , Vancomycin/administration & dosage , Vancomycin/adverse effects
12.
J Ind Microbiol Biotechnol ; 43(2-3): 195-204, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26670919

ABSTRACT

The primary challenge for treating Clostridium difficile infections (CDI) is maintenance of clinical response after the end of treatment (sustained clinical response). Disease recurrence following a positive clinical response occurs in approximately 6-25 % of patients after the first episode and in up to 65 % for subsequent recurrences. Surotomycin, a novel cyclic lipopeptide antibiotic with a core derived by Streptomyces roseosporus fermentation, disrupts C. difficile cellular membrane activity in both logarithmic and stationary phases and minimally disturbs normal gastrointestinal microbiota because of its lack of activity against Gram-negative anaerobes and facultative anaerobes. Preclinical and clinical evidence indicate that surotomycin has low oral bioavailability, allowing gastrointestinal tract concentrations to greatly exceed its minimum inhibitory concentration for C. difficile. Surotomycin is well tolerated and effective in hamster models of CDI. Phase 2 clinical evidence suggests that surotomycin (250 mg twice daily) is an effective CDI treatment, with statistically lower recurrence rates than vancomycin.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/therapeutic use , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Lipopeptides/isolation & purification , Lipopeptides/therapeutic use , Peptides, Cyclic/isolation & purification , Peptides, Cyclic/therapeutic use , Animals , Anti-Bacterial Agents/pharmacology , Biological Availability , Clinical Trials, Phase II as Topic , Clostridioides difficile/cytology , Gastrointestinal Microbiome/drug effects , Humans , Lipopeptides/administration & dosage , Lipopeptides/pharmacokinetics , Lipopeptides/pharmacology , Microbial Sensitivity Tests , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/pharmacology
13.
Antimicrob Agents Chemother ; 59(7): 4139-47, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25941217

ABSTRACT

Clostridium difficile infection (CDI) is an urgent public health concern causing considerable clinical and economic burdens. CDI can be treated with antibiotics, but recurrence of the disease following successful treatment of the initial episode often occurs. Surotomycin is a rapidly bactericidal cyclic lipopeptide antibiotic that is in clinical trials for CDI treatment and that has demonstrated superiority over vancomycin in preventing CDI relapse. Surotomycin is a structural analogue of the membrane-active antibiotic daptomycin. Previously, we utilized in vitro serial passage experiments to derive C. difficile strains with reduced surotomycin susceptibilities. The parent strains used included ATCC 700057 and clinical isolates from the restriction endonuclease analysis (REA) groups BI and K. Serial passage experiments were also performed with vancomycin-resistant and vancomycin-susceptible Enterococcus faecium and Enterococcus faecalis. The goal of this study is to identify mutations associated with reduced surotomycin susceptibility in C. difficile and enterococci. Illumina sequence data generated for the parent strains and serial passage isolates were compared. We identified nonsynonymous mutations in genes coding for cardiolipin synthase in C. difficile ATCC 700057, enoyl-(acyl carrier protein) reductase II (FabK) and cell division protein FtsH2 in C. difficile REA type BI, and a PadR family transcriptional regulator in C. difficile REA type K. Among the 4 enterococcal strain pairs, 20 mutations were identified, and those mutations overlap those associated with daptomycin resistance. These data give insight into the mechanism of action of surotomycin against C. difficile, possible mechanisms for resistance emergence during clinical use, and the potential impacts of surotomycin therapy on intestinal enterococci.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Drug Resistance, Bacterial/genetics , Enterococcus/drug effects , Enterococcus/genetics , Lipopeptides/pharmacology , Peptides, Cyclic/pharmacology , Bacterial Proteins/genetics , Computational Biology , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Daptomycin/pharmacology , Humans , Microbial Sensitivity Tests , Mutation/genetics , Prohibitins , Restriction Mapping , Vancomycin Resistance/genetics
14.
Antimicrob Agents Chemother ; 59(9): 5165-70, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26055381

ABSTRACT

Surotomycin (CB-183,315), a cyclic lipopeptide, is in phase 3 clinical development for the treatment of Clostridium difficile infection. We report here the further characterization of the in vitro mode of action of surotomycin, including its activity against growing and nongrowing C. difficile. This was assessed through time-kill kinetics, allowing a determination of the effects on the membrane potential and permeability and macromolecular synthesis in C. difficile. Against representative strains of C. difficile, surotomycin displayed concentration-dependent killing of both logarithmic-phase and stationary-phase cultures at a concentration that was ≤16× the MIC. Exposure resulted in the inhibition of macromolecular synthesis (in DNA, RNA, proteins, and cell wall). At bactericidal concentrations, surotomycin dissipated the membrane potential of C. difficile without changes to the permeability of propidium iodide. These observations are consistent with surotomycin acting as a membrane-active antibiotic, exhibiting rapid bactericidal activities against growing and nongrowing C. difficile.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Lipopeptides/pharmacology , Peptides, Cyclic/pharmacology , Clostridioides difficile/growth & development , Microbial Sensitivity Tests
15.
Antimicrob Agents Chemother ; 59(7): 4199-205, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25941230

ABSTRACT

The increasing incidence and severity of infection by Clostridium difficile have stimulated attempts to develop new antimicrobial therapies. We report here the relative abilities of two antibiotics (metronidazole and vancomycin) in current use for treating C. difficile infection and of a third antimicrobial, surotomycin, to kill C. difficile cells at various stages of development and to inhibit the production of the toxin proteins that are the major virulence factors. The results indicate that none of the drugs affects the viability of spores at 8× MIC or 80× MIC and that all of the drugs kill exponential-phase cells when provided at 8× MIC. In contrast, none of the drugs killed stationary-phase cells or inhibited toxin production when provided at 8× MIC and neither vancomycin nor metronidazole killed stationary-phase cells when provided at 80× MIC. Surotomycin, on the other hand, did kill stationary-phase cells when provided at 80× MIC but did so without inducing lysis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Toxins/biosynthesis , Clostridioides difficile/drug effects , Clostridioides difficile/metabolism , Enterotoxins/biosynthesis , Lipopeptides/pharmacology , Peptides, Cyclic/pharmacology , Bacterial Toxins/genetics , Cell Wall/drug effects , Clostridioides difficile/genetics , Enterotoxins/genetics , Gene Expression Regulation, Bacterial/drug effects , Metronidazole/pharmacology , Microbial Sensitivity Tests , Mutation/genetics , Spores, Bacterial/drug effects , Vancomycin/pharmacology , Virulence Factors/metabolism
16.
Antimicrob Agents Chemother ; 58(7): 3976-82, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24798273

ABSTRACT

Surotomycin (CB-183,315) is an orally administered, minimally absorbed, selective bactericidal cyclic lipopeptide in phase 3 development for the treatment of Clostridium difficile-associated diarrhea. The aim of this study was to evaluate the emergence of resistance in C. difficile (ATCC 700057 and three recent clinical isolates from the restriction endonuclease analysis groups BI, BK, and K), vancomycin-susceptible (VS) Enterococcus faecalis (ATCC 49452), vancomycin-resistant (VR) E. faecalis (ATCC 700802), VS Enterococcus faecium (ATCC 6569), and VR E. faecium (ATCC 51559) under anaerobic conditions. The rate of spontaneous resistance was below the limit of detection (<10(-8) to <10(-9)) for surotomycin at 16 and 32× the MIC for all isolates tested. Under selective pressure by serial passage, C. difficile grew in a maximum of 4 µg/ml surotomycin (final MICs of 2 to 8 µg/ml [4- to 16-fold higher than those of the naive control]) at day 15, with the exception of the C. difficile BK strain, which grew in 16 to 32 µg/ml (final MICs of 8 to 32 µg/ml [16- to 64-fold higher than those of the naive control]). Enterococci remained relatively unchanged over 15 days, growing in a maximum of 8 µg/ml surotomycin (final MICs of 2 to 16 µg/ml [8- to 64-fold higher than those of the naive control]). Of the isolates tested, no cross-resistance to vancomycin, rifampin, ampicillin, metronidazole, or moxifloxacin was observed. Surotomycin at 20× MIC demonstrated equally rapid bactericidal activity (≥ 3-log-unit reduction in CFU/ml in ≤ 8 h) against naive and reduced-susceptibility isolates of C. difficile, VS Enterococcus (VSE), and VR Enterococcus (VRE), except for C. difficile BK (2.6-log-unit reductions for both). These results suggest that emergence of resistance to surotomycin against C. difficile, E. faecalis, and E. faecium is likely to be rare.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Enterococcus faecalis/drug effects , Enterococcus faecium/drug effects , Lipopeptides/pharmacology , Peptides, Cyclic/pharmacology , Bacteria, Anaerobic/drug effects , Microbial Sensitivity Tests , Vancomycin Resistance
17.
PLoS Pathog ; 8(11): e1003012, 2012.
Article in English | MEDLINE | ID: mdl-23166491

ABSTRACT

The Legionella pneumophila effector protein RalF functions as a guanine nucleotide exchange factor (GEF) that activates the host small GTPase protein ADP-ribosylation factor (Arf), and recruits this host protein to the vacuoles in which this pathogen resides. GEF activity is conferred by the Sec7 domain located in the N-terminal region of RalF. Structural studies indicate that the C-terminal region of RalF makes contacts with residues in the Sec7 domain important for Arf interactions. Theoretically, the C-terminal region of RalF could prevent nucleotide exchange activity by blocking the ability of Arf to interact with the Sec7 domain. For this reason, the C-terminal region of RalF has been termed a capping domain. Here, the role of the RalF capping domain was investigated by comparing biochemical and effector activities mediated by this domain in both the Legionella RalF protein (LpRalF) and in a RalF ortholog isolated from the unrelated intracellular pathogen Rickettsia prowazekii (RpRalF). These data indicate that both RalF proteins contain a functional Sec7 domain and that the capping domain regulates RalF GEF activity. The capping domain has intrinsic determinants that mediate localization of the RalF protein inside of host cells and confer distinct effector activities. Localization mediated by the capping domain of LpRalF enables the GEF to modulate membrane transport in the secretory pathway, whereas, the capping domain of RpRalF enables this bacterial GEF to modulate actin dynamics occurring near the plasma membrane. Thus, these data reveal that divergence in the function of the C-terminal capping domain alters the in vivo functions of the RalF proteins.


Subject(s)
Bacterial Proteins/metabolism , Cell Membrane/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Legionella pneumophila/metabolism , Animals , Bacterial Proteins/genetics , CHO Cells , Cell Membrane/genetics , Cricetinae , Cricetulus , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Humans , Legionella pneumophila/genetics , Protein Binding , Protein Structure, Tertiary , Rickettsia prowazekii/genetics , Rickettsia prowazekii/metabolism
18.
World J Gastrointest Pathophysiol ; 14(4): 71-85, 2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37727283

ABSTRACT

BACKGROUND: The Centers for Disease Control and Prevention estimate that Clostridioides difficile (C. difficile) causes half a million infections (CDI) annually and is a major cause of total infectious disease death in the United States, causing inflammation of the colon and potentially deadly diarrhea. We recently reported the isolation of ADS024, a Bacillus velezensis (B. velezensis) strain, which demonstrated direct in vitro bactericidal activity against C. difficile, with minimal collateral impact on other members of the gut microbiota. In this study, we hypothesized that in vitro activities of ADS024 will translate in vivo to protect against CDI challenge in mouse models. AIM: To investigate the in vivo efficacy of B. velezensis ADS024 in protecting against CDI challenge in mouse models. METHODS: To mimic disruption of the gut microbiota, the mice were exposed to vancomycin prior to dosing with ADS024. For the mouse single-dose study, the recovery of ADS024 was assessed via microbiological analysis of intestinal and fecal samples at 4 h, 8 h, and 24 h after a single oral dose of 5 × 108 colony-forming units (CFU)/mouse of freshly grown ADS024. The single-dose study in miniature swine included groups that had been pre-dosed with vancomycin and that had been exposed to a dose range of ADS024, and a group that was not pre-dosed with vancomycin and received a single dose of ADS024. The ADS024 colonies [assessed by quantitative polymerase chain reaction (qPCR) using ADS024-specific primers] were counted on agar plates. For the 28-d miniature swine study, qPCR was used to measure ADS024 levels from fecal samples after oral administration of ADS024 capsules containing 5 × 109 CFU for 28 consecutive days, followed by MiSeq compositional sequencing and bioinformatic analyses to measure the impact of ADS024 on microbiota. Two studies were performed to determine the efficacy of ADS024 in a mouse model of CDI: Study 1 to determine the effects of fresh ADS024 culture and ADS024 spore preparations on the clinical manifestations of CDI in mice, and Study 2 to compare the efficacy of single daily doses vs dosing 3 times per day with fresh ADS024. C. difficile challenge was performed 24 h after the start of ADS024 exposure. To model the human distal colon, an anerobic fecal fermentation system was used. MiSeq compositional sequencing and bioinformatic analyses were performed to measure microbiota diversity changes following ADS024 treatment. To assess the potential of ADS024 to be a source of antibiotic resistance, its susceptibility to 18 different antibiotics was tested. RESULTS: In a mouse model of CDI challenge, single daily doses of ADS024 were as efficacious as multiple daily doses in protecting against subsequent challenge by C. difficile pathogen-induced disease. ADS024 showed no evidence of colonization based on the observation that the ADS024 colonies were not recovered 24 h after single doses in mice or 72 h after single doses in miniature swine. In a 28-d repeat-dose study in miniature swine, ADS024 was not detected in fecal samples using plating and qPCR methods. Phylogenetic analysis performed in the human distal colon model showed that ADS024 had a selective impact on the healthy human colonic microbiota, similarly to the in vivo studies performed in miniature swine. Safety assessments indicated that ADS024 was susceptible to all the antibiotics tested, while in silico testing revealed a low potential for off-target activity or virulence and antibiotic-resistance mechanisms. CONCLUSION: Our findings, demonstrating in vivo efficacy of ADS024 in protecting against CDI challenge in mouse models, support the use of ADS024 in preventing recurrent CDI following standard antibiotic treatment.

19.
Front Microbiol ; 14: 1284083, 2023.
Article in English | MEDLINE | ID: mdl-38268707

ABSTRACT

Epithelial cell apoptosis and compromised gut barrier function are features of inflammatory bowel disease. ADS024 is a single-strain live biotherapeutic product (LBP) of Bacillus velezensis under development for treating ulcerative colitis (UC). The cytoprotective effects of the sterile filtrate of ADS024's secreted products on UC patient-derived colonic tissues, human primary colonic epithelial cells (HPEC), and human colonic epithelial T84 cells were evaluated. ADS024 filtrate significantly inhibited apoptosis and inflammation with reduced Bcl-2 Associated X-protein (BAX) and tumor necrosis factor (TNF) mRNA expression in fresh colonic explants from UC patients. Exposure to UC patient-derived serum exosomes (UCSE) induced apoptosis with increased cleaved caspase 3 protein expression in HPECs. ADS024 filtrate diminished the UCSE-mediated apoptosis by inhibiting cleaved caspase 3. TNFα and interferon-gamma (IFNγ) damaged epithelial barrier integrity with reduced transepithelial electrical resistance (TEER). ADS024 filtrate partially attenuated the TEER reduction and restored tight junction protein 1 (TJP1) expression. Oral live ADS024 treatment reduced weight loss, disease activity, colonic mucosal injury, and colonic expression of interleukin 6 (IL-6) and TNFα in dextran sodium sulfate (DSS)-treated mice with colitis. Thus, ADS024 may protect the colonic epithelial barrier in UC via anti-inflammatory, anti-apoptotic, and tight-junction protection mechanisms.

20.
Front Microbiol ; 13: 1072534, 2022.
Article in English | MEDLINE | ID: mdl-36704560

ABSTRACT

Clostridioides difficile infection (CDI) causes intestinal injury. Toxin A and toxin B cause intestinal injury by inducing colonic epithelial cell apoptosis. ADS024 is a Bacillus velezensis strain in development as a single-strain live biotherapeutic product (SS-LBP) to prevent the recurrence of CDI following the completion of standard antibiotic treatment. We evaluated the protective effects of the sterile filtrate and ethyl acetate extract of conditioned media from ADS024 and DSM7 (control strain) against mucosal epithelial injury in toxin-treated human colonic tissues and apoptosis in toxin-treated human colonic epithelial cells. Ethyl acetate extracts were generated from conditioned culture media from DSM7 and ADS024. Toxin A and toxin B exposure caused epithelial injury in fresh human colonic explants. The sterile filtrate of ADS024, but not DSM7, prevented toxin B-mediated epithelial injury in fresh human colonic explants. Both sterile filtrate and ethyl acetate extract of ADS024 prevented toxin-mediated apoptosis in human colonic epithelial cells. The anti-apoptotic effects of ADS024 filtrate and ethyl acetate extract were dependent on the inhibition of caspase 3 cleavage. The sterile filtrate, but not ethyl acetate extract, of ADS024 partially degraded toxin B. ADS024 inhibits toxin B-mediated apoptosis in human colonic epithelial cells and colonic explants.

SELECTION OF CITATIONS
SEARCH DETAIL