Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Publication year range
1.
J Antimicrob Chemother ; 77(6): 1706-1709, 2022 05 29.
Article in English | MEDLINE | ID: mdl-35229156

ABSTRACT

OBJECTIVES: STM-001, a retargeted glycopeptide, is active against MDR E. coli expressing ESBLs including carbapenemases. Herein, we assessed its capability to combat E. coli complicated urinary tract infections (cUTI) in mice driven by clinically important serine (CTX-M-15) and metallo-ß-lactamases (NDM-1). METHODS: Plasma and urine pharmacokinetics following IV administration of STM-001 (1-50 mg/kg) were determined in mice via LC-MS/MS. The effects on bacterial burden (kidney, bladder and urine) were determined in a 7 day mouse cUTI model whereby STM-001 was administered q12h or q24h at 2-100 mg/kg/day from Day 4. Efficacy was assessed by the change in log10 cfu/g or log10 cfu/mL from vehicle-treated infected mice. RESULTS: MICs of STM-001 for CTX-M-15 and NDM-1 E. coli were 8 and 16 mg/L, respectively. Blood pharmacokinetic profile was linear and dose-dependent with low clearance of 9.49 ±â€Š0.31 mL/min/kg, V = 0.63 ±â€Š0.02 L/kg and t½â€Š= 1.16 ±â€Š0.03 h. High STM-001 concentrations were recovered in urine 0-8 h post-administration, reaching up to 120-fold above its MIC. In cUTI efficacy studies, STM-001 (1-50 mg/kg, q12h) reduced CTX-M-15 burden by log10 4.31 (kidney), 3.95 (bladder) and 4.82 (urine) compared with vehicle-treated animals (P < 0.0001). STM-001 also reduced NDM-1 burden by log10 3.89 (kidney), 3.76 (bladder) and 3.08 (urine) (P < 0.0001), with similar inhibitory effects following q24h dosing. CONCLUSIONS: STM-001 was highly effective in reducing E. coli burden in kidney, bladder and urine in mouse cUTI models. The observed efficacy with either dosing regimen indicates potential low humanized doses of 1-5 mg/kg. These data support further development of STM-001 as an innovative, carbapenem-sparing antibiotic to combat human cUTIs.


Subject(s)
Carbapenem-Resistant Enterobacteriaceae , Escherichia coli Infections , Urinary Tract Infections , Animals , Anti-Bacterial Agents/pharmacology , Arginine/pharmacology , Arginine/therapeutic use , Carbapenems/pharmacology , Carbapenems/therapeutic use , Chromatography, Liquid , Escherichia coli , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Mice , Microbial Sensitivity Tests , Tandem Mass Spectrometry , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology , Vancomycin/pharmacology , beta-Lactamases/pharmacology
2.
Article in English | MEDLINE | ID: mdl-33468474

ABSTRACT

The ability of vancomycin-arginine (V-r) to extend the spectrum of activity of glycopeptides to Gram-negative bacteria was investigated. Its MIC towards Escherichia coli, including ß-lactamase expressing Ambler classes A, B, and D, was 8 to 16 µg/ml. Addition of 8 times the MIC of V-r to E. coli was acutely bactericidal and associated with a low frequency of resistance (<2.32 × 10-10). In vivo, V-r markedly reduced E. coli burden by >7 log10 CFU/g in a thigh muscle model. These data warrant further development of V-r in combatting E. coli, including resistant forms.


Subject(s)
Escherichia coli , Vancomycin , Anti-Bacterial Agents/pharmacology , Arginine , Escherichia coli/genetics , Microbial Sensitivity Tests , Vancomycin/pharmacology
3.
Article in English | MEDLINE | ID: mdl-32253212

ABSTRACT

Third-generation cephalosporin (3GC)-resistant Enterobacteriaceae are classified as critical priority pathogens, with extended-spectrum ß-lactamases (ESBLs) as principal resistance determinants. Enmetazobactam (formerly AAI101) is a novel ESBL inhibitor developed in combination with cefepime for empirical treatment of serious Gram-negative infections in settings where ESBLs are prevalent. Cefepime-enmetazobactam has been investigated in a phase 3 trial in patients with complicated urinary tract infections or acute pyelonephritis. This study examined pharmacokinetic-pharmacodynamic (PK-PD) relationships of enmetazobactam, in combination with cefepime, for ESBL-producing isolates of Klebsiella pneumoniae in 26-h murine neutropenic thigh infection models. Enmetazobactam dose fractionation identified the time above a free threshold concentration (fT > CT ) as the PK-PD index predictive of efficacy. Nine ESBL-producing isolates of K. pneumoniae, resistant to cefepime and piperacillin-tazobactam, were included in enmetazobactam dose-ranging studies. The isolates encoded CTX-M-type, SHV-12, DHA-1, and OXA-48 ß-lactamases and covered a cefepime-enmetazobactam MIC range from 0.06 to 2 µg/ml. Enmetazobactam restored the efficacy of cefepime against all isolates tested. Sigmoid curve fitting across the combined set of isolates identified enmetazobactam PK-PD targets for stasis and for a 1-log10 bioburden reduction of 8% and 44% fT > 2 µg/ml, respectively, with a concomitant cefepime PK-PD target of 40 to 60% fT > cefepime-enmetazobactam MIC. These findings support clinical dose selection and breakpoint setting for cefepime-enmetazobactam.


Subject(s)
Cephalosporins , Thigh , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Azabicyclo Compounds , Cefepime , Humans , Klebsiella pneumoniae , Mice , Microbial Sensitivity Tests , Triazoles , beta-Lactamases/genetics
4.
J Antimicrob Chemother ; 75(10): 2919-2924, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32719853

ABSTRACT

BACKGROUND: During antifungal prophylaxis, micafungin is generally infused IV once daily over 1 h. In practice, less-frequent dosing could improve the quality of life in patients requiring long-term treatment or prophylaxis. The feasibility of this approach was assessed using humanized doses of daily or infrequent micafungin regimens. OBJECTIVES: To evaluate the effectiveness of intermittent high-dose micafungin, simulating human exposure, for prophylaxis of invasive candidiasis in a rat model of chronic Candida albicans gastrointestinal colonization and systemic dissemination. METHODS: Two weeks post-infection with an oral challenge of C. albicans, Sprague-Dawley rats were immunocompromised with a cytotoxic drug and a steroid. Rats received IV infusions of: daily vehicle control; daily subcutaneous micafungin (20 mg/kg SC); high-dose micafungin (20 mg/kg bolus SC + 80 mg/kg infusion/72 h, to simulate intermittent human dosing of 300 mg/72 h); or daily fluconazole by mouth (10 mg/kg PO). The effects of antifungal prophylaxis on faecal fungal burden and systemic C. albicans dissemination were evaluated. RESULTS: A rat model of chronic C. albicans gastrointestinal colonization and systemic dissemination was established, characterized by a sustained microbiological burden over 29 days and fungal recovery from normally sterile tissues. Using this model, intermittent high-dose micafungin (delivered via iPrecio pumps) to simulate humanized doses of 300 mg/72 h was significantly more effective than vehicle control, as effective as once-daily micafungin and similar to daily fluconazole at reducing faecal burden and preventing systemic dissemination. CONCLUSIONS: These data indicate that intermittent high-dose micafungin can be as effective as daily therapy, supporting clinical assessment in high-risk patients requiring long-term antifungal prophylaxis.


Subject(s)
Candida albicans , Echinocandins , Animals , Antifungal Agents/therapeutic use , Humans , Lipopeptides , Micafungin , Quality of Life , Rats , Rats, Sprague-Dawley
5.
Article in English | MEDLINE | ID: mdl-29530861

ABSTRACT

Infections caused by carbapenem-resistant Enterobacteriaceae (CRE) are increasingly prevalent and have become a major worldwide threat to human health. Carbapenem resistance is driven primarily by the acquisition of ß-lactamase enzymes, which are able to degrade carbapenem antibiotics (hence termed carbapenemases) and result in high levels of resistance and treatment failure. Clinically relevant carbapenemases include both serine ß-lactamases (SBLs; e.g., KPC-2 and OXA-48) and metallo-ß-lactamases (MBLs), such as NDM-1. MBL-producing strains are endemic within the community in many Asian countries, have successfully spread worldwide, and account for many significant CRE outbreaks. Recently approved combinations of ß-lactam antibiotics with ß-lactamase inhibitors are active only against SBL-producing pathogens. Therefore, new drugs that specifically target MBLs and which restore carbapenem efficacy against MBL-producing CRE pathogens are urgently needed. Here we report the discovery of a novel MBL inhibitor, ANT431, that can potentiate the activity of meropenem (MEM) against a broad range of MBL-producing CRE and restore its efficacy against an Escherichia coli NDM-1-producing strain in a murine thigh infection model. This is a strong starting point for a chemistry lead optimization program that could deliver a first-in-class MBL inhibitor-carbapenem combination. This would complement the existing weaponry against CRE and address an important and growing unmet medical need.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carbapenem-Resistant Enterobacteriaceae/drug effects , Meropenem/pharmacology , Microbial Sensitivity Tests , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/genetics
6.
J Antimicrob Chemother ; 73(11): 3068-3073, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30351438

ABSTRACT

Objectives: In vitro and in vivo activity of the dihydroorotate dehydrogenase inhibitor olorofim (formerly F901318) (F2G Limited, UK) against clinically relevant species of the Aspergillus section Terrei was evaluated. Methods: A total of 92 clinical Aspergillus section Terrei isolates [42 Aspergillus terreus sensu stricto and 50 cryptic species: Aspergillus alabamensis (n = 8), Aspergillus citrinoterreus (n = 27), Aspergillus floccosus (n = 1), Aspergillus hortai (n = 13) and Aspergillus neoafricanus (n = 1)] were evaluated. MICs were determined using the CLSI M38-A2 method. MICs of olorofim were compared with those of posaconazole, voriconazole, itraconazole and amphotericin B. The in vivo efficacy of olorofim was determined in an immunosuppressed murine model of disseminated aspergillosis. Results: Olorofim was highly active against all tested Aspergillus section Terrei isolates, exhibiting an MIC range of 0.002-0.063 mg/L. Slightly higher MICs were observed for A. terreus cryptic species. Olorofim MICs were lower than those observed for the azoles. Selected strains with elevated MICs of azoles were highly susceptible to olorofim. Olorofim administered by oral and intravenous routes produced survival rates of 90%-100% in A. terreus-infected mice. Conclusions: Olorofim showed potent and consistent in vitro activity against all A. terreus strains tested, including those with elevated MICs of other antifungal substances. Overall, growth inhibition by olorofim was superior to that of azoles. In vivo data showed that olorofim was highly efficacious in prolonging survival of mice with disseminated aspergillosis due to A. terreus sensu stricto.


Subject(s)
Acetamides/pharmacology , Acetamides/therapeutic use , Antifungal Agents/pharmacology , Aspergillosis/drug therapy , Aspergillus/drug effects , Invasive Fungal Infections/drug therapy , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Piperazines/pharmacology , Piperazines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use , Animals , Dihydroorotate Dehydrogenase , Disease Models, Animal , Immunocompromised Host , Male , Mice , Mice, Inbred ICR , Microbial Sensitivity Tests
7.
Article in English | MEDLINE | ID: mdl-28533232

ABSTRACT

Novel approaches for the treatment of multidrug-resistant Gram-negative bacterial infections are urgently required. One approach is to potentiate the efficacy of existing antibiotics whose spectrum of activity is limited by the permeability barrier presented by the Gram-negative outer membrane. Cationic peptides derived from polymyxin B have been used to permeabilize the outer membrane, granting antibiotics that would otherwise be excluded access to their targets. We assessed the in vitro efficacies of combinations of SPR741 with conventional antibiotics against Escherichia coli, Klebsiella pneumoniae, and Acinetobacter baumannii Of 35 antibiotics tested, the MICs of 8 of them were reduced 32- to 8,000-fold against E. coli and K. pneumoniae in the presence of SPR741. The eight antibiotics, azithromycin, clarithromycin, erythromycin, fusidic acid, mupirocin, retapamulin, rifampin, and telithromycin, had diverse targets and mechanisms of action. Against A. baumannii, similar potentiation was achieved with clarithromycin, erythromycin, fusidic acid, retapamulin, and rifampin. Susceptibility testing of the most effective antibiotic-SPR741 combinations was extended to 25 additional multidrug-resistant or clinical isolates of E. coli and K. pneumoniae and 17 additional A. baumannii isolates in order to rank the potentiated antibiotics. SPR741 was also able to potentiate antibiotics that are substrates of the AcrAB-TolC efflux pump in E. coli, effectively circumventing the contribution of this pump to intrinsic antibiotic resistance. These studies support the further development of SPR741 in combination with conventional antibiotics for the treatment of Gram-negative bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Negative Bacterial Infections/drug therapy , Polymyxin B/pharmacology , Acinetobacter baumannii/drug effects , Antimicrobial Cationic Peptides/chemistry , Cell Membrane Permeability/drug effects , Drug Synergism , Escherichia coli/drug effects , Gram-Negative Bacterial Infections/microbiology , Klebsiella pneumoniae/drug effects , Microbial Sensitivity Tests
8.
J Antimicrob Chemother ; 72(3): 778-781, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27999015

ABSTRACT

Objectives: To investigate the efficacy of a potent novel antimicrobial protein of mass 6 kDa, epidermicin NI01, for eradicating the nasal burden of MRSA in a cotton rat ( Sigmodon hispidus ) model. Methods: MRSA strain ATCC 43300 was used to establish a robust colonization of cotton rat nares. This model was used to evaluate the efficacy of topical 0.04% and 0.2% epidermicin NI01, administered twice daily for 3 days consecutively, and topical 0.8% epidermicin NI01 administered once, for reducing nasal MRSA burden. Control groups remained untreated or were administered vehicle only (0.5% hydroxypropylmethylcellulose) or 2% mupirocin twice daily for 3 days. The experiment was terminated at day 5 and MRSA quantitative counts were determined. Tissues recovered from animals treated with 0.2% epidermicin twice daily for 3 days were examined for histological changes. Results: Mupirocin treatment resulted in a reduction in burden of log 10 (log R) of 2.59 cfu/nares compared with vehicle ( P < 0.0001). Epidermicin NI01 administered once at 0.8% showed excellent efficacy, resulting in a log R of 2.10 cfu/nares ( P = 0.0004), which was equivalent to mupirocin. Epidermicin NI01 administered at 0.2% or 0.04% twice daily for 3 days did not have a significant impact on the tissue burden recovered from the nares. Mild to marked histological abnormalities were noted, but these were determined to be reversible. Conclusion: A single dose of topical epidermicin NI01 was as effective as mupirocin administered twice daily for 3 days in eradication of MRSA from the nares of cotton rats. This justifies further development of epidermicin for this indication.


Subject(s)
Antimicrobial Cationic Peptides/administration & dosage , Bacteriocins/administration & dosage , Methicillin-Resistant Staphylococcus aureus/drug effects , Nose/microbiology , Staphylococcal Infections/drug therapy , Administration, Topical , Animals , Antimicrobial Cationic Peptides/therapeutic use , Bacterial Load/drug effects , Bacteriocins/therapeutic use , Dose-Response Relationship, Drug , Mupirocin/therapeutic use , Nose/drug effects , Rats , Sigmodontinae , Staphylococcal Infections/microbiology
9.
Antimicrob Agents Chemother ; 60(11): 6471-6482, 2016 11.
Article in English | MEDLINE | ID: mdl-27527088

ABSTRACT

Clostridium difficile causes infections of the colon in susceptible patients. Specifically, gut dysbiosis induced by treatment with broad-spectrum antibiotics facilitates germination of ingested C. difficile spores, expansion of vegetative cells, and production of symptom-causing toxins TcdA and TcdB. The current standard of care for C. difficile infections (CDI) consists of administration of antibiotics such as vancomycin that target the bacterium but also perpetuate gut dysbiosis, often leading to disease recurrence. The monoclonal antitoxin antibodies actoxumab (anti-TcdA) and bezlotoxumab (anti-TcdB) are currently in development for the prevention of recurrent CDI. In this study, the effects of vancomycin or actoxumab/bezlotoxumab treatment on progression and resolution of CDI were assessed in mice and hamsters. Rodent models of CDI are characterized by an early severe phase of symptomatic disease, associated with high rates of morbidity and mortality; high intestinal C. difficile burden; and a disrupted intestinal microbiota. This is followed in surviving animals by gradual recovery of the gut microbiota, associated with clearance of C. difficile and resolution of disease symptoms over time. Treatment with vancomycin prevents disease initially by inhibiting outgrowth of C. difficile but also delays microbiota recovery, leading to disease relapse following discontinuation of therapy. In contrast, actoxumab/bezlotoxumab treatment does not impact the C. difficile burden but rather prevents the appearance of toxin-dependent symptoms during the early severe phase of disease, effectively preventing disease until the microbiota (the body's natural defense against C. difficile) has fully recovered. These data provide insight into the mechanism of recurrence following vancomycin administration and into the mechanism of recurrence prevention observed clinically with actoxumab/bezlotoxumab.


Subject(s)
Anti-Bacterial Agents/adverse effects , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Antitoxins/pharmacology , Clostridium Infections/drug therapy , Vancomycin/adverse effects , Animals , Anti-Bacterial Agents/administration & dosage , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/biosynthesis , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/biosynthesis , Broadly Neutralizing Antibodies , Clostridioides difficile/drug effects , Clostridioides difficile/growth & development , Clostridioides difficile/pathogenicity , Clostridium Infections/immunology , Clostridium Infections/microbiology , Clostridium Infections/mortality , Convalescence , Cricetulus , Disease Models, Animal , Disease Progression , Enterotoxins/antagonists & inhibitors , Enterotoxins/biosynthesis , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Humans , Mice , Mice, Inbred C57BL , Survival Analysis , Vancomycin/administration & dosage
10.
J Antimicrob Chemother ; 70(6): 1757-62, 2015.
Article in English | MEDLINE | ID: mdl-25652749

ABSTRACT

OBJECTIVES: SMT19969 is a novel narrow-spectrum antimicrobial under development for the treatment of Clostridium difficile infection (CDI). The objectives were to assess the relative efficacies of SMT19969, vancomycin and fidaxomicin in the hamster model of CDI. METHODS: Hamsters were infected with either C. difficile BI1 (ribotype 027) or C. difficile 630 (ribotype 012) prior to treatment with vehicle, SMT19969, fidaxomicin or vancomycin for 5 days. Animals were further monitored through to day 28 and survival recorded. Plasma and gastrointestinal concentrations of SMT19969 following single and repeat administration in infected hamsters were determined. RESULTS: Following infection with C. difficile BI1, treatment with SMT19969, vancomycin and fidaxomicin resulted in 100% survival during the 5 day dosing period, with 90%-100% of animals receiving SMT19969 and fidaxomicin surviving during the post-dosing follow-up period. Whilst protective during treatment, onset of mortality was observed on day 11 in animals treated with vancomycin, with a 10% survival recorded by day 28. Similar results were observed for SMT19969 and vancomycin following infection with C. difficile 630, with day 28 survival rates of 80%-100% and 0%, respectively. Fidaxomicin protected animals infected with C. difficile 630 from mortality during dosing, although day 28 survival rates varied from 0% to 40% depending on dose. Plasma levels of SMT19969 were typically below the limit of quantification, but levels in the gastrointestinal tract remained far in excess of the MIC. CONCLUSIONS: These data show that SMT19969 is highly effective at treating both acute infection and preventing recurrent disease and support continued investigation of SMT19969 as a potential therapy for CDI.


Subject(s)
Aminoglycosides/administration & dosage , Anti-Bacterial Agents/administration & dosage , Benzimidazoles/administration & dosage , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Pyridines/administration & dosage , Vancomycin/administration & dosage , Animals , Anti-Bacterial Agents/pharmacokinetics , Benzimidazoles/pharmacokinetics , Clostridium Infections/microbiology , Disease Models, Animal , Fidaxomicin , Gastrointestinal Tract/chemistry , Mesocricetus , Microbial Sensitivity Tests , Plasma/chemistry , Pyridines/pharmacokinetics , Recurrence , Survival Analysis , Treatment Outcome
11.
Ann Neurol ; 75(5): 670-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24644058

ABSTRACT

OBJECTIVE: Bacterial infection contributes to diverse noninfectious diseases and worsens outcome after stroke. Streptococcus pneumoniae, the most common infection in patients at risk of stroke, is a major cause of prolonged hospitalization and death of stroke patients, but how infection impacts clinical outcome is not known. METHODS: We induced sustained pulmonary infection by a human S. pneumoniae isolate in naive and comorbid rodents to investigate the effect of infection on vascular and inflammatory responses prior to and after cerebral ischemia. RESULTS: S. pneumoniae infection triggered atherogenesis, led to systemic induction of interleukin (IL) 1, and profoundly exacerbated (50-90%) ischemic brain injury in rats and mice, a response that was more severe in combination with old age and atherosclerosis. Systemic blockade of IL-1 with IL-1 receptor antagonist (IL-1Ra) fully reversed infection-induced exacerbation of brain injury and functional impairment caused by cerebral ischemia. We show that infection-induced systemic inflammation mediates its effects via increasing platelet activation and microvascular coagulation in the brain after cerebral ischemia, as confirmed by reduced brain injury in response to blockade of platelet glycoprotein (GP) Ibα. IL-1 and platelet-mediated signals converge on microglia, as both IL-1Ra and GPIbα blockade reversed the production of IL-1α by microglia in response to cerebral ischemia in infected animals. INTERPRETATION: S. pneumoniae infection augments atherosclerosis and exacerbates ischemic brain injury via IL-1 and platelet-mediated systemic inflammation. These mechanisms may contribute to diverse cardio- and cerebrovascular pathologies in humans.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Interleukin-1/adverse effects , Platelet Glycoprotein GPIb-IX Complex/adverse effects , Streptococcal Infections/metabolism , Streptococcal Infections/pathology , Streptococcus pneumoniae , Animals , Brain Ischemia/microbiology , Disease Progression , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Interleukin-1/physiology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microglia/microbiology , Microglia/pathology , Platelet Activation , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/physiology , Rats , Rats, Wistar , Streptococcal Infections/microbiology
12.
Antimicrob Agents Chemother ; 57(6): 2793-800, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23571544

ABSTRACT

Fluconazole is frequently the only antifungal agent that is available for induction therapy for cryptococcal meningitis. There is relatively little understanding of the pharmacokinetics and pharmacodynamics (PK-PD) of fluconazole in this setting. PK-PD relationships were estimated with 4 clinical isolates of Cryptococcus neoformans. MICs were determined using Clinical and Laboratory Standards Institute (CLSI) methodology. A nonimmunosuppressed murine model of cryptococcal meningitis was used. Mice received two different doses of fluconazole (125 mg/kg of body weight/day and 250 mg/kg of body weight/day) orally for 9 days; a control group of mice was not given fluconazole. Fluconazole concentrations in plasma and in the cerebrum were determined using high-performance liquid chromatography (HPLC). The cryptococcal density in the brain was estimated using quantitative cultures. A mathematical model was fitted to the PK-PD data. The experimental results were extrapolated to humans (bridging study). The PK were linear. A dose-dependent decline in fungal burden was observed, with near-maximal activity evident with dosages of 250 mg/kg/day. The MIC was important for understanding the exposure-response relationships. The mean AUC/MIC ratio associated with stasis was 389. The results of the bridging study suggested that only 66.7% of patients receiving 1,200 mg/kg would achieve or exceed an AUC/MIC ratio of 389. The potential breakpoints for fluconazole against Cryptococcus neoformans follow: susceptible, ≤ 2 mg/liter; resistant, >2 mg/liter. Fluconazole may be an inferior agent for induction therapy because many patients cannot achieve the pharmacodynamic target. Clinical breakpoints are likely to be significantly lower than epidemiological cutoff values. The MIC may guide the appropriate use of fluconazole. If fluconazole is the only option for induction therapy, then the highest possible dose should be used.


Subject(s)
Antifungal Agents/pharmacokinetics , Antifungal Agents/therapeutic use , Cryptococcus neoformans/drug effects , Fluconazole/pharmacokinetics , Fluconazole/therapeutic use , Meningitis, Cryptococcal/drug therapy , Meningoencephalitis/drug therapy , Animals , Antifungal Agents/administration & dosage , Antifungal Agents/pharmacology , Area Under Curve , Disease Models, Animal , Fluconazole/administration & dosage , Fluconazole/pharmacology , Humans , Male , Meningitis, Cryptococcal/microbiology , Meningoencephalitis/microbiology , Mice , Microbial Sensitivity Tests/standards , Models, Biological , Treatment Outcome
13.
J Antimicrob Chemother ; 68(11): 2569-75, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23800902

ABSTRACT

OBJECTIVES: To evaluate the in vivo effectiveness of a combination treatment containing ranalexin (a natural antimicrobial peptide) and lysostaphin (an antistaphylococcal endopeptidase) for reducing nasal burden of methicillin-resistant Staphylococcus aureus (MRSA). METHODS: The community-acquired MRSA strain S. aureus NRS384 (USA300-0114) was used in the present study because it is commonly isolated from human nares and it established consistent and reproducible colonization of cotton rat nares. This model was used to evaluate the efficacy of ranalexin/lysostaphin gels (0.1%-1% w/v; administered intranasally once or once per day for 3 consecutive days) for reducing nasal MRSA burden. Control animals were administered vehicle gel only (0.5% hydroxypropyl methylcellulose) or 2% mupirocin, which is used clinically for nasal decolonization of MRSA. Nasal MRSA burden was assessed at 192 h post-inoculation, which was at least 72 h after the final treatment had been administered. An additional study assessed the efficacy of 0.1% ranalexin/lysostaphin against a mupirocin-resistant MRSA strain (MUP20), which had been selected by serial passage of S. aureus NRS384 through subinhibitory concentrations of mupirocin. RESULTS: Gels containing 0.1% ranalexin/lysostaphin consistently reduced median nasal burden of MRSA to an extent similar to or greater than 2% mupirocin. Treatment with 0.1% ranalexin/lysostaphin was also effective against the MUP20 strain. There was evidence for only minimal irritancy in cotton rat nares administered three doses of 0.1% ranalexin/lysostaphin, suggesting that this agent is suitable for short-course therapy such as is employed currently for nasal decolonization with mupirocin. CONCLUSIONS: Ranalexin/lysostaphin could serve as an alternative to mupirocin for nasal decolonization of MRSA.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Carrier State/drug therapy , Lysostaphin/administration & dosage , Methicillin-Resistant Staphylococcus aureus/drug effects , Nose/microbiology , Peptides, Cyclic/administration & dosage , Staphylococcal Infections/drug therapy , Administration, Topical , Animals , Bacterial Load , Carrier State/microbiology , Drug Therapy, Combination/methods , Gels/administration & dosage , Models, Animal , Sigmodontinae , Staphylococcal Infections/microbiology , Treatment Outcome
14.
J Infect Dis ; 206(3): 442-52, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22634880

ABSTRACT

BACKGROUND: Voriconazole is a first-line agent for the treatment of invasive pulmonary aspergillosis (IPA). There are increasing reports of Aspergillus fumigatus isolates with reduced susceptibility to voriconazole. METHODS: An in vitro dynamic model of IPA was developed that enabled simulation of human-like voriconazole pharmacokinetics. Galactomannan was used as a biomarker. The pharmacodynamics of voriconazole against wild-type and 3 resistant strains of A. fumigatus were defined. The results were bridged to humans to provide decision support for setting breakpoints for voriconazole using Clinical Laboratory Standards Institute (CLSI) and European Committee of Antimicrobial Susceptibility Testing (EUCAST) methodologies. RESULTS: Isolates with higher minimum inhibitory concentrations (MICs) required higher area under the concentration time curves (AUCs) to achieve suppression of galactomannan. Using CLSI and EUCAST methodologies, the AUC:MIC values that achieved suppression of galactomannan were 55 and 32.1, respectively. Using CLSI and EUCAST methodologies, the trough concentration:MIC values that achieved suppression of galactomannan were 1.68 and 1, respectively. Potential CLSI breakpoints for voriconazole are ≤ 0.5 mg/L for susceptible and >1 mg/L for resistant. Potential EUCAST breakpoints for voriconazole are ≤1 mg/L for susceptible and >2 mg/L for resistant. CONCLUSIONS: This dynamic model of IPA is a useful tool to address many remaining questions related to antifungal treatment of Aspergillus spp.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus fumigatus/drug effects , Invasive Pulmonary Aspergillosis/drug therapy , Pyrimidines/pharmacology , Triazoles/pharmacology , Antifungal Agents/pharmacokinetics , Aspergillus fumigatus/metabolism , Bioreactors , Cell Culture Techniques , Cells, Cultured , Chromatography, High Pressure Liquid , Computer Simulation , Decision Support Techniques , Dose-Response Relationship, Drug , Drug Monitoring , Drug Resistance, Fungal , Endothelial Cells/cytology , Endothelial Cells/microbiology , Epithelial Cells/cytology , Galactose/analogs & derivatives , Humans , Mannans/metabolism , Microbial Sensitivity Tests , Models, Biological , Pulmonary Artery/cytology , Pyrimidines/pharmacokinetics , Respiratory Mucosa/cytology , Triazoles/pharmacokinetics , Voriconazole
15.
Antimicrob Agents Chemother ; 56(10): 5180-5, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22825124

ABSTRACT

Voriconazole is a first-line agent for the treatment of invasive pulmonary aspergillosis. Isolates with elevated voriconazole MICs are increasingly being seen, and the optimal treatment regimen is not defined. We investigated whether the combination of voriconazole with anidulafungin may be beneficial for the treatment of A. fumigatus strains with elevated voriconazole MICs. We used an in vitro model of the human alveolus to define the exposure-response relationships for a wild-type strain (voriconazole MIC, 0.5 mg/liter) and strains with defined molecular mechanisms of triazole resistance (MICs, 4 to 16 mg/liter). All strains had anidulafungin minimum effective concentrations (MECs) of 0.0078 mg/liter. Exposure-response relationships were estimated using galactomannan as a biomarker. Concentrations of voriconazole and anidulafungin were measured using high-performance liquid chromatography (HPLC). The interaction of voriconazole and anidulafungin was described using the Greco model. Fungal growth was progressively inhibited with higher drug exposures of voriconazole. Strains with elevated voriconazole MICs required proportionally greater voriconazole exposures to achieve a comparable antifungal effect. Galactomannan concentrations were only marginally reduced by anidulafungin monotherapy. An additive effect between voriconazole and anidulafungin was apparent. In conclusion, the addition of anidulafungin does not markedly alter the exposure-response relationship of voriconazole. A rise in serum galactomannan during combination therapy with voriconazole and anidulafungin should be interpreted as treatment failure and not attributed to a paradoxical reaction related to echinocandin treatment.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus fumigatus/drug effects , Echinocandins/pharmacology , Invasive Pulmonary Aspergillosis/microbiology , Pyrimidines/pharmacology , Triazoles/pharmacology , Anidulafungin , Antifungal Agents/pharmacokinetics , Cell Line , Chromatography, High Pressure Liquid , Drug Interactions , Echinocandins/pharmacokinetics , Humans , Microbial Sensitivity Tests , Models, Theoretical , Pulmonary Alveoli , Pyrimidines/pharmacokinetics , Triazoles/pharmacokinetics , Voriconazole
16.
Antimicrob Agents Chemother ; 56(2): 708-14, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22123680

ABSTRACT

Hematogenous Candida meningoencephalitis (HCME) is a serious infection in premature neonates. Anidulafungin is an echinocandin antifungal agent with potent activity against Candida spp., but its efficacy and optimal regimens for human neonates with HCME are not known. A well-validated rabbit model of HCME was used to define pharmacokinetic-pharmacodynamic (PK-PD) relationships of anidulafungin. A mathematical model was fitted to the entire data set. The experimental data were bridged to humans. A population PK model was fitted to the data from human neonates receiving anidulafungin receiving a loading dose of 3 mg/kg, followed by 1.5 mg/kg/day. Monte Carlo simulations were performed to identify candidate anidulafungin regimens for humans. All untreated rabbits succumbed by ≤96 h postinoculation. The PK of anidulafungin was linear with dose-dependent penetration into the cerebrum. Anidulafungin exerted a rapid antifungal effect that was apparent in the first dosing interval. Near-maximal antifungal activity was observed with dosages of 10 to 20 mg/kg/day. The bridging studies suggested that the current regimen of first 3 mg/kg, followed by 1.5 mg/kg/day, is suboptimal. Higher dosages were associated with progressively greater antifungal effect. Anidulafungin is effective for the treatment of experimental HCME. Higher dosages than those currently used for clinical care are required for maximal antifungal effect.


Subject(s)
Antifungal Agents , Candida albicans/drug effects , Candidiasis/drug therapy , Disease Models, Animal , Echinocandins , Infant, Premature, Diseases/drug therapy , Meningoencephalitis/drug therapy , Anidulafungin , Animals , Antifungal Agents/pharmacokinetics , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Area Under Curve , Candidiasis/microbiology , Echinocandins/administration & dosage , Echinocandins/pharmacokinetics , Echinocandins/pharmacology , Echinocandins/therapeutic use , Humans , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/microbiology , Male , Meningitis, Fungal/drug therapy , Meningitis, Fungal/microbiology , Meningoencephalitis/microbiology , Monte Carlo Method , Rabbits , Treatment Outcome
17.
Antimicrob Agents Chemother ; 56(8): 4146-53, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22615280

ABSTRACT

Itraconazole is used for the prevention and treatment of infections caused by Aspergillus fumigatus. An understanding of the pharmacodynamics of itraconazole against wild-type and triazole-resistant strains provides a basis for innovative therapeutic strategies for treatment of infections. An in vitro model of the human alveolus was used to define the pharmacodynamics of itraconazole. Galactomannan was used as a biomarker. The effect of systemic and airway administration of itraconazole was assessed, as was a combination of itraconazole administered to the airway and systemically administered 5FC. Systemically administered itraconazole against the wild type induced a concentration-dependent decline in galactomannan in the alveolar and endothelial compartments. No exposure-response relationships were apparent for the L98H, M220T, or G138C mutant. The administration of itraconazole to the airway resulted in comparable exposure-response relationships to those observed with systemic therapy. This was achieved without detectable concentrations of drug within the endothelial compartment. The airway administration of itraconazole resulted in a definite but submaximal effect in the endothelial compartment against the L98H mutant. The administration of 5FC resulted in a concentration-dependent decline in galactomannan in both the alveolar and endothelial compartments. The combination of airway administration of itraconazole and systemically administered 5FC was additive. Systemic administration of itraconazole is ineffective against Cyp51 mutants. The airway administration of itraconazole is effective for the treatment of wild-type strains and appears to have some activity against the L98H mutants. Combination with other agents, such as 5FC, may enable the attainment of near-maximal antifungal activity.


Subject(s)
Antifungal Agents/pharmacology , Aspergillosis/drug therapy , Aspergillus fumigatus/drug effects , Itraconazole/pharmacology , Lung Diseases, Fungal/drug therapy , Pulmonary Alveoli/microbiology , Antifungal Agents/administration & dosage , Antifungal Agents/pharmacokinetics , Aspergillosis/microbiology , Aspergillosis/prevention & control , Cells, Cultured , Drug Administration Routes , Drug Resistance, Fungal , Flucytosine/administration & dosage , Flucytosine/pharmacology , Galactose/analogs & derivatives , Humans , Itraconazole/administration & dosage , Itraconazole/pharmacokinetics , Lung Diseases, Fungal/microbiology , Mannans/analysis , Microbial Sensitivity Tests , Triazoles/pharmacology
18.
J Infect Dis ; 203(9): 1324-32, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21357943

ABSTRACT

BACKGROUND: Posaconazole is a triazole with anti-Aspergillus activity. However, little is known about the utility of posaconazole as primary therapy for invasive pulmonary aspergillosis. METHODS: An in vitro model of the human alveolus was used to study the impact of minimum inhibitory concentrations (MIC) on exposure-response relationships. The pharmacokinetic-pharmacodynamic relationships of posaconazole were examined in an inhalational murine model of invasive pulmonary aspergillosis. A mathematical model was fitted to the entire data set. This model was then used to describe the relationship between drug exposure, quantified in terms of the area under the concentration time curve to MIC (AUC:MIC) and the observed antifungal effect. RESULTS: The posaconazole MIC was an important determinant of exposure-response relationships and accounted for a portion of the observed variance. Murine pharmacokinetics were linear for dosages 1-20 mg/kg/day. There was a dose-dependent decline in serum galactomannan concentrations, with near-maximal suppression following 20 mg/kg/day. The murine pharmacokinetic-pharmacodynamic data were well described by the mathematical model. An AUC:MIC ratio of 167 was associated with half-maximal antifungal effect. CONCLUSIONS: These results provide the experimental foundation for the selection of candidate posaconazole regimens for the primary treatment of invasive pulmonary aspergillosis in profoundly neutropenic hosts.


Subject(s)
Antifungal Agents/administration & dosage , Antifungal Agents/pharmacokinetics , Invasive Pulmonary Aspergillosis/microbiology , Triazoles/administration & dosage , Triazoles/pharmacokinetics , Animals , Disease Models, Animal , Invasive Pulmonary Aspergillosis/drug therapy , Male , Mice , Models, Theoretical
19.
Antimicrob Agents Chemother ; 55(5): 2092-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21321135

ABSTRACT

This study investigated the phase-dependent expression and activity of efflux pumps in Aspergillus fumigatus treated with voriconazole. Fourteen strains were shown to become increasingly resistant in the 12-h (16- to 128-fold) and 24-h (>512-fold) phases compared to 8-h germlings. An Ala-Nap uptake assay demonstrated a significant increase in efflux pump activity in the 12-h and 24-h phases (P<0.0001). The efflux pump activity of the 8-h germling cells was also significantly induced by voriconazole (P<0.001) after 24 h of treatment. Inhibition of efflux pump activity with the competitive substrate MC-207,110 reduced the voriconazole MIC values for the A. fumigatus germling cells by 2- to 8-fold. Quantitative expression analysis of AfuMDR4 mRNA transcripts showed a phase-dependent increase as the mycelial complexity increased, which was coincidental with a strain-dependent increase in azole resistance. Voriconazole also significantly induced this in a time-dependent manner (P<0.001). Finally, an in vivo mouse biofilm model was used to evaluate efflux pump expression, and it was shown that AfuMDR4 was constitutively expressed and significantly induced by treatment with voriconazole after 24 h (P<0.01). Our results demonstrate that efflux pumps are expressed in complex A. fumigatus biofilm populations and that this contributes to azole resistance. Moreover, voriconazole treatment induces efflux pump expression. Collectively, these data may provide evidence for azole treatment failures in clinical cases of aspergillosis.


Subject(s)
Aspergillus fumigatus/drug effects , Aspergillus fumigatus/metabolism , Azoles/pharmacology , Biofilms/drug effects , Fungal Proteins/metabolism , Animals , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Aspergillosis/drug therapy , Aspergillus fumigatus/genetics , Drug Resistance, Fungal/genetics , Fungal Proteins/genetics , Male , Mice , Microbial Sensitivity Tests , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Triazoles/pharmacology , Triazoles/therapeutic use , Voriconazole
20.
Antimicrob Agents Chemother ; 55(11): 5113-21, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21876055

ABSTRACT

Multiple Aspergillus fumigatus isolates from a patient with two aspergillomas complicating chronic pulmonary aspergillosis were pan-azole resistant. Microsatellite typing was identical for all isolates despite major phenotypic and some growth rate differences. Three different cyp51A mutations were found (G138C, Y431C, and G434C), of which the first two were demonstrated by heterologous expression in a hypersusceptible Saccharomyces cerevisiae strain to be at least partly responsible for elevated MICs. cyp51A and cyp51B gene duplication was excluded, but increased expression of cyp51A was demonstrated in three isolates selected for additional study (7-to 13-fold increases). In the isolate with the greatest cyp51A expression, an Aft1 transposon was found inserted 370 bp upstream of the start codon of the cyp51A gene, an integration location never previously demonstrated in Aspergillus. Two transcription start sites were identified at 49 and 136 bp upstream of the start codon. The role of the Aft1 transposon, if any, in modulating cyp51A expression remains to be established. Increased mRNA expression of the transporters AfuMDR1 and AfuMDR4 also was demonstrated in some isolates, which could contribute to azole resistance or simply represent a stress response. The diversity of confirmed and possible azole resistance mechanisms demonstrated in a single series of isogenic isolates is remarkable, indicating the ability of A. fumigatus to adapt in the clinical setting.


Subject(s)
Aspergillus fumigatus/drug effects , Aspergillus fumigatus/genetics , Azoles/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Fungal Proteins/metabolism , Polymorphism, Single Nucleotide/genetics , Antifungal Agents/pharmacology , Aspergillus fumigatus/metabolism , Cytochrome P-450 Enzyme System/genetics , DNA Transposable Elements/genetics , Drug Resistance, Fungal/genetics , Fungal Proteins/genetics , Promoter Regions, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL