Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Antimicrob Agents Chemother ; 67(4): e0221321, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36946741

ABSTRACT

Omadacycline, a novel aminomethylcycline with in vitro activity against Gram-positive and -negative organisms, including Streptococcus pneumoniae and Haemophilus influenzae, is approved in the United States to treat patients with community-acquired bacterial pneumonia (CABP). Using nonclinical pharmacokinetic-pharmacodynamic (PK-PD) targets for efficacy and in vitro surveillance data for omadacycline against S. pneumoniae and H. influenzae, and a population pharmacokinetic model, PK-PD target attainment analyses were undertaken using total-drug epithelial lining fluid (ELF) and free-drug plasma exposures to evaluate omadacycline 100 mg intravenously (i.v.) every 12 h or 200 mg i.v. every 24 h (q24h) on day 1, followed by 100 mg i.v. q24h on day 2 and 300 mg orally q24h on days 3 to 5 for patients with CABP. Percent probabilities of PK-PD target attainment on days 1 and 2 by MIC were assessed using the following four approaches for selecting PK-PD targets: (i) median, (ii) second highest, (iii) highest, and (iv) randomly assigned total-drug ELF and free-drug plasma ratio of the area under the concentration-time curve to the MIC (AUC/MIC ratio) targets associated with a 1-log10 CFU reduction from baseline. Percent probabilities of PK-PD target attainment based on total-drug ELF AUC/MIC ratio targets on days 1 and 2 were ≥91.1% for S. pneumoniae for all approaches but the highest target and ≥99.2% for H. influenzae for all approaches at MIC90s (0.12 and 1 µg/mL for S. pneumoniae and H. influenzae, respectively). Lower percent probabilities of PK-PD target attainment based on free-drug plasma AUC/MIC ratio targets were observed for randomly assigned and the highest free-drug plasma targets for S. pneumoniae and for all targets for H. influenzae. These data provided support for approved omadacycline dosing regimens to treat patients with CABP and decisions for the interpretive criteria for the in vitro susceptibility testing of omadacycline against these pathogens.


Subject(s)
Pneumonia, Bacterial , Streptococcus pneumoniae , Humans , Anti-Bacterial Agents/pharmacology , Bacteria , Haemophilus influenzae , Microbial Sensitivity Tests , Pneumonia, Bacterial/drug therapy
2.
Int J Infect Dis ; 104: 501-509, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33484864

ABSTRACT

OBJECTIVE: Community-acquired bacterial pneumonia (CABP) is a major clinical burden worldwide. In the phase III OPTIC study (NCT02531438) in CABP, omadacycline was found to be non-inferior to moxifloxacin for investigator-assessed clinical response (IACR) at post-treatment evaluation (PTE, 5-10 days after last dose). This article reports the efficacy findings, as specified in the European Medicines Agency (EMA) guidance. METHODS: Patients were randomized 1:1 to omadacycline 100 mg intravenously (every 12 h for two doses, then every 24 h) with optional transition to 300 mg orally after 3 days, or moxifloxacin 400 mg intravenously (every 24 h) with optional transition to 400 mg orally after 3 days. The total treatment duration was 7-14 days. The primary endpoint for EMA efficacy analysis was IACR at PTE in patients with Pneumonia Patient Outcomes Research Team (PORT) risk class III and IV. RESULTS: In total, 660 patients were randomized as PORT risk class III and IV. Omadacycline was non-inferior to moxifloxacin at PTE. The clinical success rates were 88.4% and 85.2%, respectively [intent-to-treat population; difference 3.3; 97.5% confidence interval (CI) -2.7 to 9.3], and 92.5% and 90.5%, respectively (clinically evaluable population; difference 2.0; 97.5% CI 3.2-7.4). Clinical success rates with omadacycline and moxifloxacin were similar against identified pathogens and across key subgroups. CONCLUSIONS: Omadacycline was non-inferior to moxifloxacin for IACR at PTE, with high clinical success across pathogen types and patient subgroups.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Community-Acquired Infections/drug therapy , Moxifloxacin/therapeutic use , Pneumonia, Bacterial/drug therapy , Tetracyclines/therapeutic use , Administration, Intravenous , Aged , Anti-Bacterial Agents/administration & dosage , Community-Acquired Infections/microbiology , Double-Blind Method , Female , Humans , Male , Middle Aged , Moxifloxacin/administration & dosage , Pneumonia, Bacterial/microbiology , Tetracyclines/administration & dosage
3.
Article in English | MEDLINE | ID: mdl-32340986

ABSTRACT

Omadacycline, a novel aminomethylcycline antibiotic with activity against Gram-positive and -negative organisms, including tetracycline-resistant pathogens, received FDA approval in October 2018 for the treatment of patients with acute bacterial skin and skin structure infections (ABSSSI) and community-acquired bacterial pneumonia (CABP). A previously developed population pharmacokinetic (PK) model based on phase 1 intravenous and oral PK data was refined using data from infected patients. Data from 10 phase 1 studies used to develop the previous model were pooled with data from three additional phase 1 studies, a phase 1b uncomplicated urinary tract infection study, one phase 3 CABP study, and two phase 3 ABSSSI studies. The final population PK model was a three-compartment model with first-order absorption using transit compartments to account for absorption delay following oral dosing and first-order elimination. Epithelial lining fluid (ELF) concentrations were modeled as a subcompartment of the first peripheral compartment. A food effect on oral bioavailability was included in the model. Sex was the only significant covariate identified, with 15.6% lower clearance for females than males. Goodness-of-fit diagnostics indicated a precise and unbiased fit to the data. The final model, which was robust in its ability to predict plasma and ELF exposures following omadacycline administration, was also able to predict the central tendency and variability in concentration-time profiles using an external phase 3 ABSSSI data set. A population PK model, which described omadacycline PK in healthy subjects and infected patients, was developed and subsequently used to support pharmacokinetic-pharmacodynamic (PK-PD) and PK-PD target attainment assessments.


Subject(s)
Community-Acquired Infections , Tetracyclines , Administration, Intravenous , Anti-Bacterial Agents/therapeutic use , Clinical Trials, Phase I as Topic , Clinical Trials, Phase III as Topic , Community-Acquired Infections/drug therapy , Female , Humans , Male , Tetracyclines/therapeutic use
4.
Article in English | MEDLINE | ID: mdl-32284378

ABSTRACT

Omadacycline is a novel aminomethylcycline with activity against Gram-positive and -negative organisms, including Haemophilus influenzae, which is one of the leading causes of community-acquired bacterial pneumonia (CABP). The evaluation of antimicrobial agents against H. influenzae using standard murine infection models is challenging due to the low pathogenicity of this species in mice. Therefore, 24-h dose-ranging studies using a one-compartment in vitro infection model were undertaken with the goal of characterizing the magnitude of the ratio of the area under the concentration-time curve (AUC) to the MIC (AUC/MIC ratio) associated with efficacy for a panel of five H. influenzae isolates. These five isolates, for which MIC values were 1 or 2 mg/liter, were exposed to omadacycline total-drug epithelial lining fluid (ELF) concentration-time profiles based on those observed in healthy volunteers following intravenous omadacycline administration. Relationships between change in log10 CFU/ml from baseline at 24 h and the total-drug ELF AUC/MIC ratios for each isolate and for the isolates pooled were evaluated using Hill-type models and nonlinear least-squares regression. As evidenced by the high coefficients of determination (r2) of 0.88 to 0.98, total-drug ELF AUC/MIC ratio described the data well for each isolate and the isolates pooled. The median total-drug ELF AUC/MIC ratios associated with net bacterial stasis and 1- and 2-log10 CFU/ml reductions from baseline at 24 h were 6.91, 8.91, and 11.1, respectively. These data were useful to support the omadacycline dosing regimens selected for the treatment of patients with CABP, as well as susceptibility breakpoints for H. influenzae.


Subject(s)
Haemophilus influenzae , Streptococcus pneumoniae , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Humans , Mice , Microbial Sensitivity Tests , Tetracyclines
5.
Lancet Infect Dis ; 19(10): 1080-1090, 2019 10.
Article in English | MEDLINE | ID: mdl-31474458

ABSTRACT

BACKGROUND: Pathogen resistance and safety concerns limit oral antibiotic options for the treatment of acute bacterial skin and skin structure infections (ABSSSI). We aimed to compare the efficacy and safety of once-daily oral omadacycline, an aminomethylcycline antibiotic, versus twice-daily oral linezolid for treatment of ABSSSI. METHODS: In this phase 3, double-blind, randomised, non-inferiority study, eligible adults with ABSSSI at 33 sites in the USA were randomly assigned (1:1) to receive omadacycline (450 mg orally every 24 h over the first 48 h then 300 mg orally every 24 h) or linezolid (600 mg orally every 12 h) for 7-14 days. Randomisation was done via an interactive response system using a computer-generated schedule, and stratified by type of infection (wound infection, cellulitis or erysipelas, or major abscess) and receipt (yes or no) of allowed previous antibacterial treatment. Investigators, funders, and patients were masked to treatment assignments. Primary endpoints were early clinical response, 48-72 h after first dose, in the modified intention-to-treat (mITT) population (randomised patients without solely Gram-negative ABSSSI pathogens at baseline), and investigator-assessed clinical response at post-treatment evaluation, 7-14 days after the last dose, in the mITT population and clinically evaluable population (ie, mITT patients who had a qualifying infection as per study-entry criteria, received study drug, did not receive a confounding antibiotic, and had an assessment of outcome during the protocol-defined window). The safety population included randomised patients who received any amount of study drug. We set a non-inferiority margin of 10%. This study is registered with ClinicalTrials.gov, NCT02877927, and is complete. FINDINGS: Between Aug 11, 2016, and June 6, 2017, 861 participants were assessed for eligibility. 735 participants were randomly assigned, of whom 368 received omadacycline and 367 received linezolid. Omadacycline (315 [88%] of 360) was non-inferior to linezolid (297 [83%] of 360) for early clinical response (percentage-point difference 5·0, 95% CI -0·2 to 10·3) in the mITT population. For investigator-assessed clinical response at post-treatment evaluation, omadacycline was non-inferior to linezolid in the mITT (303 [84%] of 360 vs 291 [81%] of 360; percentage-point difference 3·3, 95% CI -2·2 to 9·0) and clinically evaluable (278 [98%] of 284 vs 279 [96%] of 292; 2·3, -0·5 to 5·8) populations. Mild to moderate nausea and vomiting were the most frequent treatment-emergent adverse events in omadacycline (111 [30%] of 368 and 62 [17%] of 368, respectively) and linezolid (28 [8%] of 367 and 11 [3%] of 367, respectively) groups. INTERPRETATION: Once-daily oral omadacycline was non-inferior to twice-daily oral linezolid in adults with ABSSSI, and was safe and well tolerated. Oral-only omadacycline represents a new treatment option for ABSSSI, with potential for reduction in hospital admissions and cost savings. FUNDING: Paratek Pharmaceuticals.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Linezolid/administration & dosage , Linezolid/therapeutic use , Skin Diseases, Bacterial/drug therapy , Tetracyclines/administration & dosage , Tetracyclines/therapeutic use , Administration, Oral , Adult , Anti-Bacterial Agents/adverse effects , Double-Blind Method , Female , Humans , Length of Stay , Linezolid/adverse effects , Male , Middle Aged , Nausea/etiology , Tetracyclines/adverse effects , Treatment Outcome , Vomiting/etiology
6.
N Engl J Med ; 380(6): 528-538, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30726689

ABSTRACT

BACKGROUND: Acute bacterial skin and skin-structure infections are associated with substantial morbidity and health care costs. Omadacycline, an aminomethylcycline antibiotic that can be administered once daily either orally or intravenously, is active against pathogens that commonly cause such infections, including antibiotic-resistant strains. METHODS: In this double-blind trial, we randomly assigned adults with acute bacterial skin and skin-structure infections (in a 1:1 ratio) to receive omadacycline (100 mg given intravenously every 12 hours for two doses, then 100 mg given intravenously every 24 hours) or linezolid (600 mg given intravenously every 12 hours). A transition to oral omadacycline (300 mg every 24 hours) or oral linezolid (600 mg every 12 hours) was allowed after 3 days; the total treatment duration was 7 to 14 days. The primary end point was an early clinical response at 48 to 72 hours, defined as survival with a reduction in lesion size of at least 20% without rescue antibacterial therapy. A secondary end point was an investigator-assessed clinical response at the post-treatment evaluation 7 to 14 days after the last dose, with clinical response defined as survival with resolution or improvement in signs or symptoms of infection to the extent that further antibacterial therapy was unnecessary. For both end points, the noninferiority margin was 10 percentage points. RESULTS: In the modified intention-to-treat population, omadacycline (316 patients) was noninferior to linezolid (311 patients) with respect to early clinical response (rate of response, 84.8% and 85.5%, respectively; difference, -0.7 percentage points; 95% confidence interval [CI], -6.3 to 4.9). Omadacycline also was noninferior to linezolid with respect to investigator-assessed clinical response at the post-treatment evaluation in the modified intention-to-treat population (rate of response, 86.1% and 83.6%, respectively; difference, 2.5 percentage points; 95% CI, -3.2 to 8.2) and in the clinical per-protocol population (96.3% and 93.5%, respectively; difference, 2.8 percentage points; 95% CI, -1.0 to 6.9). In both groups, the efficacy of the trial drug was similar for methicillin-susceptible and methicillin-resistant Staphylococcus aureus infections. Adverse events were reported in 48.3% of the patients in the omadacycline group and in 45.7% of those in the linezolid group; the most frequent adverse events in both groups were gastrointestinal (in 18.0% and 15.8% of the patients in the respective groups). CONCLUSIONS: Omadacycline was noninferior to linezolid for the treatment of acute bacterial skin and skin-structure infections and had a similar safety profile. (Funded by Paratek Pharmaceuticals; OASIS-1 ClinicalTrials.gov number, NCT02378480 .).


Subject(s)
Anti-Bacterial Agents/therapeutic use , Linezolid/therapeutic use , Skin Diseases, Bacterial/drug therapy , Tetracyclines/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Anti-Bacterial Agents/adverse effects , Double-Blind Method , Drug Administration Schedule , Drug Resistance, Bacterial , Female , Humans , Infusions, Intravenous , Intention to Treat Analysis , Linezolid/adverse effects , Male , Methicillin-Resistant Staphylococcus aureus/drug effects , Middle Aged , Skin Diseases, Bacterial/microbiology , Tetracyclines/adverse effects , Young Adult
7.
N Engl J Med ; 380(6): 517-527, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30726692

ABSTRACT

BACKGROUND: Omadacycline, a new once-daily aminomethylcycline antibiotic agent that can be administered intravenously or orally, reaches high concentrations in pulmonary tissues and is active against common pathogens that cause community-acquired bacterial pneumonia. METHODS: In a double-blind trial, we randomly assigned (in a 1:1 ratio) adults with community-acquired bacterial pneumonia (Pneumonia Severity Index risk class II, III, or IV) to receive omadacycline (100 mg intravenously every 12 hours for two doses, then 100 mg intravenously every 24 hours), or moxifloxacin (400 mg intravenously every 24 hours). A transition to oral omadacycline (300 mg every 24 hours) or moxifloxacin (400 mg every 24 hours), respectively, was allowed after 3 days; the total treatment duration was 7 to 14 days. The primary end point was early clinical response, defined as survival with improvement in at least two of four symptoms (cough, sputum production, pleuritic chest pain, and dyspnea) and no worsening of symptoms at 72 to 120 hours, without receipt of rescue antibacterial therapy. A secondary end point was investigator-assessed clinical response at a post-treatment evaluation 5 to 10 days after the last dose, with clinical response defined as resolution or improvement in signs or symptoms to the extent that further antibacterial therapy was unnecessary. A noninferiority margin of 10 percentage points was used. RESULTS: The intention-to-treat population included 386 patients in the omadacycline group and 388 patients in the moxifloxacin group. Omadacycline was noninferior to moxifloxacin for early clinical response (81.1% and 82.7%, respectively; difference, -1.6 percentage points; 95% confidence interval [CI], -7.1 to 3.8), and the rates of investigator-assessed clinical response at the post-treatment evaluation were 87.6% and 85.1%, respectively (difference, 2.5 percentage points; 95% CI, -2.4 to 7.4). Adverse events that emerged after treatment initiation were reported in 41.1% of the patients in the omadacycline group and 48.5% of the patients in the moxifloxacin group; the most frequent events were gastrointestinal (10.2% and 18.0%, respectively), and the largest difference was for diarrhea (1.0% and 8.0%). Twelve deaths (8 in the omadacycline group and 4 in the moxifloxacin group) occurred during the trial. CONCLUSIONS: Omadacycline was noninferior to moxifloxacin for the treatment of community-acquired bacterial pneumonia in adults. (Funded by Paratek Pharmaceuticals; OPTIC ClinicalTrials.gov number, NCT02531438 .).


Subject(s)
Anti-Bacterial Agents/therapeutic use , Moxifloxacin/therapeutic use , Pneumonia, Bacterial/drug therapy , Tetracyclines/therapeutic use , Adult , Aged , Aged, 80 and over , Anti-Bacterial Agents/adverse effects , Bacteria/isolation & purification , Community-Acquired Infections/drug therapy , Double-Blind Method , Drug Administration Schedule , Female , Hospitalization , Humans , Infusions, Intravenous , Intention to Treat Analysis , Male , Middle Aged , Moxifloxacin/adverse effects , Pneumonia, Bacterial/microbiology , Tetracyclines/adverse effects
8.
Int J Antimicrob Agents ; 49(1): 25-30, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27931793

ABSTRACT

The proliferation of multidrug-resistant Gram-negative pathogens has been exacerbated by a lack of novel agents in current development by pharmaceutical companies. Ceftolozane/tazobactam was recently approved by the FDA for the treatment of complicated intra-abdominal infections and complicated urinary tract infections. In the present study, the activity of ceftolozane/tazobactam against four isogenic Escherichia coli strains was investigated in a hollow-fibre infection model simulating various clinical dosing regimens. The four investigational E. coli strains included #2805 (no ß-lactamase), #2890 (AmpC ß-lactamase), #2842 (CMY-10 ß-lactamase) and #2807 (CTX-M-15 ß-lactamase). Each strain was exposed to regimens simulating 1 g ceftolozane, 2 g ceftolozane, 1 g ceftolozane/0.5 g tazobactam, and 2 g ceftolozane/1 g tazobactam utilising a starting inoculum of ca. 106 CFU/mL. Whereas 1 g of ceftolozane eradicated strains #2805 and #2842 without subsequent regrowth, 1 g ceftolozane/0.5 g tazobactam was required to eradicate strain #2890. For strain #2890, ceftolozane monotherapy led to bacterial growth on plates impregnated with 20 mg/L ceftolozane by 24 h, whilst combination treatment with tazobactam completely suppressed the development of ceftolozane resistance. In contrast, none of the regimens, including 2 g ceftolozane/1 g tazobactam, were able to entirely suppress bacterial growth in strain #2807, with bacterial counts exceeding 108 CFU/mL by 48 h and ceftolozane-resistant populations being amplified after 24 h. Thus, the combination of ceftolozane and tazobactam achieved bactericidal activity followed by sustained killing over 10 days for three of four isogenic E. coli strains. Ceftolozane/tazobactam is a promising new agent to counter multidrug-resistant Gram-negative bacteria.


Subject(s)
Anti-Infective Agents, Urinary/pharmacokinetics , Cephalosporins/pharmacokinetics , Escherichia coli/drug effects , Microfluidics/methods , Penicillanic Acid/analogs & derivatives , beta-Lactamase Inhibitors/pharmacokinetics , beta-Lactamases/metabolism , Anti-Infective Agents, Urinary/administration & dosage , Anti-Infective Agents, Urinary/pharmacology , Cephalosporins/administration & dosage , Cephalosporins/pharmacology , Escherichia coli/enzymology , Escherichia coli/growth & development , Humans , Microbial Viability/drug effects , Models, Biological , Models, Theoretical , Penicillanic Acid/administration & dosage , Penicillanic Acid/pharmacokinetics , Penicillanic Acid/pharmacology , Tazobactam , beta-Lactamase Inhibitors/administration & dosage , beta-Lactamase Inhibitors/pharmacology
9.
J Antimicrob Chemother ; 72(1): 268-272, 2017 01.
Article in English | MEDLINE | ID: mdl-27707990

ABSTRACT

OBJECTIVES: The increase in infections caused by drug-resistant ESBL-producing Enterobacteriaceae (ESBL-ENT) is a global concern. The characteristics and outcomes of patients infected with ESBL-ENT were examined in a pooled analysis of Phase 3 clinical trials of ceftolozane/tazobactam in patients with complicated urinary tract infections (ASPECT-cUTI) and complicated intra-abdominal infections (ASPECT-cIAI). METHODS: Trials were randomized and double blind. The ASPECT-cUTI regimen was 7 days of either intravenous ceftolozane/tazobactam (1.5 g) every 8 h or levofloxacin (750 mg) once daily. The ASPECT-cIAI regimen was 4-14 days of either intravenous ceftolozane/tazobactam (1.5 g) plus metronidazole (500 mg) or meropenem (1 g) every 8 h. Baseline cultures were obtained in both indications. Enterobacteriaceae were selected for ESBL characterization based on predefined criteria and were verified genotypically. Outcomes were assessed at the test-of-cure visit 5-9 days post-therapy in ASPECT-cUTI and 24-32 days post-randomization in ASPECT-cIAI among microbiologically evaluable (ME) patients. RESULTS: Of 2076 patients randomized, 1346 were included in the pooled ME population and 150 of 1346 (11.1%) had ESBL-ENT at baseline. At US FDA/EUCAST breakpoints of ≤2/≤1 mg/L, 81.8%/72.3% of ESBL-ENT (ESBL-Escherichia coli, 95%/88.1%; ESBL-Klebsiella pneumoniae, 56.7%/36.7%) were susceptible to ceftolozane/tazobactam versus 25.3%/24.1% susceptible to levofloxacin and 98.3%/98.3% susceptible to meropenem at CLSI/EUCAST breakpoints. Clinical cure rates for ME patients with ESBL-ENT were 97.4% (76/78) for ceftolozane/tazobactam [ESBL-E. coli, 98.0% (49 of 50); ESBL-K. pneumoniae, 94.4% (17 of 18)], 82.6% (38 of 46) for levofloxacin and 88.5% (23 of 26) for meropenem. CONCLUSIONS: Randomized trial data demonstrated high clinical cure rates with ceftolozane/tazobactam treatment of cIAI and cUTI caused by ESBL-ENT.


Subject(s)
Anti-Infective Agents, Urinary/administration & dosage , Cephalosporins/administration & dosage , Escherichia coli Infections/drug therapy , Intraabdominal Infections/drug therapy , Klebsiella Infections/drug therapy , Penicillanic Acid/analogs & derivatives , Urinary Tract Infections/drug therapy , beta-Lactamase Inhibitors/administration & dosage , Administration, Intravenous , Adolescent , Adult , Aged , Aged, 80 and over , Double-Blind Method , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/genetics , Female , Genotype , Humans , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/enzymology , Klebsiella pneumoniae/genetics , Levofloxacin/administration & dosage , Male , Meropenem , Metronidazole/administration & dosage , Middle Aged , Penicillanic Acid/administration & dosage , Tazobactam , Thienamycins/administration & dosage , Treatment Outcome , Young Adult , beta-Lactamases/genetics , beta-Lactamases/metabolism
10.
BMC Infect Dis ; 16(1): 710, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27887579

ABSTRACT

BACKGROUND: Fluoroquinolones are a guideline-recommended therapy for complicated urinary tract infections, including pyelonephritis. Elevated drug concentrations of fluoroquinolones in the urine and therapy with high-dose levofloxacin are believed to overcome resistance and effectively treat infections caused by resistant bacteria. The ASPECT-cUTI phase 3 clinical trial (ClinicalTrials.gov, NCT01345929 and NCT01345955 , both registered April 28, 2011) provided an opportunity to test this hypothesis by examining the clinical and microbiological outcomes of high-dose levofloxacin treatment by levofloxacin minimum inhibitory concentration. METHODS: Patients were randomly assigned 1:1 to ceftolozane/tazobactam (1.5 g intravenous every 8 h) or levofloxacin (750 mg intravenous once daily) for 7 days of therapy. The ASPECT-cUTI study provided data on 370 patients with at least one isolate of Enterobacteriaceae at baseline who were treated with levofloxacin. Outcomes were assessed at the test-of-cure (5-9 days after treatment) and late follow-up (21-42 days after treatment) visits in the microbiologically evaluable population (N = 327). RESULTS: Test-of-cure clinical cure rates above 90% were observed at minimum inhibitory concentrations ≤4 µg/mL. Microbiological eradication rates were consistently >90% at levofloxacin minimum inhibitory concentrations ≤0.06 µg/mL. Lack of eradication of causative pathogens at the test-of-cure visit increased the likelihood of relapse by the late follow-up visit. CONCLUSIONS: Results from this study do not support levofloxacin therapy for complicated urinary tract infections caused by organisms with levofloxacin minimum inhibitory concentrations ≥4 µg/mL. TRIAL REGISTRATION: ClinicalTrials.gov, NCT01345929 and NCT01345955.


Subject(s)
Anti-Infective Agents, Urinary/administration & dosage , Cephalosporins/administration & dosage , Enterobacteriaceae Infections/drug therapy , Levofloxacin/administration & dosage , Penicillanic Acid/analogs & derivatives , Urinary Tract Infections/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Anti-Infective Agents, Urinary/therapeutic use , Cephalosporins/therapeutic use , Dose-Response Relationship, Drug , Drug Administration Schedule , Enterobacteriaceae Infections/microbiology , Female , Follow-Up Studies , Humans , Injections, Intravenous , Levofloxacin/therapeutic use , Male , Microbial Sensitivity Tests , Middle Aged , Penicillanic Acid/administration & dosage , Penicillanic Acid/therapeutic use , Recurrence , Tazobactam , Treatment Outcome , Urinary Tract Infections/microbiology , Young Adult
11.
Antibiotics (Basel) ; 5(4)2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27669321

ABSTRACT

Omadacycline is an aminomethylcycline antibiotic with potent activity against many Gram-positive and Gram-negative pathogens, including strains carrying the major efflux and ribosome protection resistance determinants. This makes it a promising candidate for therapy of severe infectious diseases. Omadacycline inhibits bacterial protein biosynthesis and competes with tetracycline for binding to the ribosome. Its interactions with the 70S ribosome were, therefore, analyzed in great detail and compared with tigecycline and tetracycline. All three antibiotics are inhibited by mutations in the 16S rRNA that mediate resistance to tetracycline in Brachyspira hyodysenteriae, Helicobacter pylori, Mycoplasma hominis, and Propionibacterium acnes. Chemical probing with dimethyl sulfate and Fenton cleavage with iron(II)-complexes of the tetracycline derivatives revealed that each antibiotic interacts in an idiosyncratic manner with the ribosome. X-ray crystallography had previously revealed one primary binding site for tetracycline on the ribosome and up to five secondary sites. All tetracyclines analyzed here interact with the primary site and tetracycline also with two secondary sites. In addition, each derivative displays a unique set of non-specific interactions with the 16S rRNA.

13.
Antimicrob Agents Chemother ; 60(7): 4387-90, 2016 07.
Article in English | MEDLINE | ID: mdl-27139477

ABSTRACT

Ceftolozane-tazobactam is active against Gram-negative pathogens, including multidrug-resistant Pseudomonas aeruginosa In a subgroup analysis of patients with complicated intra-abdominal infections (cIAIs) involving P. aeruginosa from a phase 3 program, ceftolozane-tazobactam demonstrated potent in vitro activity against P. aeruginosa Clinical cure in the microbiologically evaluable population was 100% (26/26) for ceftolozane-tazobactam plus metronidazole and 93.1% (27/29) for meropenem. These findings support the use of ceftolozane-tazobactam in the management of cIAI when P. aeruginosa is suspected or confirmed. (This study has been registered at ClinicalTrials.gov under registration no. NCT01445665 and NCT01445678.).


Subject(s)
Anti-Bacterial Agents/therapeutic use , Cephalosporins/therapeutic use , Intraabdominal Infections/drug therapy , Penicillanic Acid/analogs & derivatives , Pseudomonas aeruginosa/pathogenicity , Adult , Aged , Double-Blind Method , Female , Humans , Male , Metronidazole/therapeutic use , Middle Aged , Penicillanic Acid/therapeutic use , Pseudomonas aeruginosa/drug effects , Tazobactam
14.
J Antimicrob Chemother ; 71(7): 2014-21, 2016 07.
Article in English | MEDLINE | ID: mdl-26994090

ABSTRACT

OBJECTIVES: Empirical fluoroquinolone therapy is widely used in treating complicated urinary tract infections (cUTIs), even in areas of high fluoroquinolone resistance. While it is believed that high antibiotic concentrations in urine might be sufficient to overcome and effectively treat infections caused by resistant bacteria, clinical trial data validating this assumption are limited. This post hoc analysis evaluated the efficacy of ceftolozane/tazobactam versus levofloxacin in the subgroup of patients with cUTIs caused by levofloxacin-resistant pathogens in a randomized, controlled trial (NCT01345929/NCT01345955). METHODS: Hospitalized adults with cUTI/pyelonephritis were randomized to 7 days of 1.5 g of ceftolozane/tazobactam every 8 h or 750 mg of levofloxacin once daily, before availability of culture and susceptibility data. A composite of microbiological eradication and clinical cure 5 to 9 days post-therapy was assessed in the microbiological modified ITT (mMITT; n = 800) and microbiologically evaluable (ME; n = 694) populations. RESULTS: In the mMITT population, there were 212 patients (26.5%) with at least one baseline uropathogen that was resistant to levofloxacin. The majority of uropathogens in this subgroup were Enterobacteriaceae (n = 186) that were susceptible to ceftolozane/tazobactam [MIC ≤2 mg/L, 88.7% (165/186)]. Among patients with levofloxacin-resistant pathogens, ceftolozane/tazobactam demonstrated significantly higher composite cure rates than levofloxacin in both the mMITT [60.0% (60/100) versus 39.3% (44/112); 95% CI for the treatment difference, 7.2%-33.2%] and ME [64.0% (57/89) versus 43.4% (43/99); 95% CI for the treatment difference, 6.3%-33.7%] populations, respectively. CONCLUSIONS: High urinary levels of levofloxacin did not reliably cure cUTIs. Seven day treatment with ceftolozane/tazobactam was more effective than high-dose levofloxacin treatment in patients with cUTI caused by levofloxacin-resistant bacteria, and it may be an alternative treatment in settings of increased fluoroquinolone resistance.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Cephalosporins/therapeutic use , Drug Resistance, Bacterial , Enterobacteriaceae Infections/drug therapy , Levofloxacin/therapeutic use , Penicillanic Acid/analogs & derivatives , Urinary Tract Infections/drug therapy , beta-Lactamase Inhibitors/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Double-Blind Method , Female , Humans , Male , Microbial Sensitivity Tests , Middle Aged , Penicillanic Acid/therapeutic use , Tazobactam , Treatment Outcome , Young Adult
15.
Antimicrob Agents Chemother ; 60(4): 2075-80, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26787689

ABSTRACT

We have previously demonstrated the pharmacokinetic-pharmacodynamic (PK-PD) index best associated with the efficacy of tazobactam when used in combination with ceftolozane to be the percentage of the dosing interval during which tazobactam concentrations remained above a threshold value (%time>threshold). Using anin vitroinfection model and the same isogenic CTX-M-15-producingEscherichia colitriplet set genetically engineered to transcribe different levels ofblaCTX-M-15, herein we describe dose fractionation studies designed to evaluate the PK-PD index associated with tazobactam efficacy, when given in combination with piperacillin, and the impact of the presence of a different ß-lactam agent, or differentblaCTX-M-15transcription levels, on the magnitude of the tazobactam PK-PD index necessary for efficacy. The recombinant strains demonstrated piperacillin MIC values of 128, >256, and >256 µg/ml for the low-, moderate-, and high-level CTX-M-15-producingE. colistrains, respectively. The MIC value for piperacillin in the presence of 4 µg/ml of tazobactam was 2 µg/ml for all three strains. The PK-PD index associated with tazobactam efficacy was confirmed to be %time>threshold, regardless of ß-lactamase transcription (r(2)= 0.839). The tazobactam concentration thresholds, however, changed with the CTX-M-15 transcription level and were 0.25, 0.5, and 2 µg/ml for the low-, moderate-, and high-level CTX-M-15-producing strains, respectively (r(2)= 0.921, 0.773, and 0.875, respectively). The %time>threshold values for tazobactam necessary for net bacterial stasis and a 1- and 2-log10-unit CFU/ml decrease from baseline at 24 h were 44.9, 62.9, and 84.9%, respectively. In addition to verifying our previous study results, these results also demonstrated that the magnitude of bacterial-cell killing associated with a ß-lactam-ß-lactamase inhibitor combination is dependent on the amount of ß-lactamase produced. These data provide important information for the development of ß-lactam-ß-lactamase inhibitor combination agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Models, Statistical , Penicillanic Acid/analogs & derivatives , Piperacillin/pharmacology , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/genetics , Anti-Bacterial Agents/pharmacokinetics , Computer Simulation , Drug Therapy, Combination , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Microbial Sensitivity Tests , Organisms, Genetically Modified , Penicillanic Acid/pharmacokinetics , Penicillanic Acid/pharmacology , Piperacillin/pharmacokinetics , Plasmids/chemistry , Plasmids/metabolism , Tazobactam , Transcription, Genetic , beta-Lactamase Inhibitors/pharmacokinetics , beta-Lactamases/metabolism
16.
Antimicrob Agents Chemother ; 60(4): 1967-73, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26729494

ABSTRACT

Despite a dearth of new agents currently being developed to combat multidrug-resistant Gram-negative pathogens, the combination of ceftolozane and tazobactam was recently approved by the Food and Drug Administration to treat complicated intra-abdominal and urinary tract infections. To characterize the activity of the combination product, time-kill studies were conducted against 4 strains ofEscherichia colithat differed in the type of ß-lactamase they expressed. The four investigational strains included 2805 (no ß-lactamase), 2890 (AmpC ß-lactamase), 2842 (CMY-10 ß-lactamase), and 2807 (CTX-M-15 ß-lactamase), with MICs to ceftolozane of 0.25, 4, 8, and >128 mg/liter with no tazobactam, and MICs of 0.25, 1, 4, and 8 mg/liter with 4 mg/liter tazobactam, respectively. All four strains were exposed to a 6 by 5 array of ceftolozane (0, 1, 4, 16, 64, and 256 mg/liter) and tazobactam (0, 1, 4, 16, and 64 mg/liter) over 48 h using starting inocula of 10(6)and 10(8)CFU/ml. While ceftolozane-tazobactam achieved bactericidal activity against all 4 strains, the concentrations of ceftolozane and tazobactam required for a ≥3-log reduction varied between the two starting inocula and the 4 strains. At both inocula, the Hill plots (R(2)> 0.882) of ceftolozane revealed significantly higher 50% effective concentrations (EC50s) at tazobactam concentrations of ≤4 mg/liter than those at concentrations of ≥16 mg/liter (P< 0.01). Moreover, the EC50s at 10(8)CFU/ml were 2.81 to 66.5 times greater than the EC50s at 10(6)CFU/ml (median, 10.7-fold increase;P= 0.002). These promising results indicate that ceftolozane-tazobactam achieves bactericidal activity against a wide range of ß-lactamase-producingE. colistrains.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Cephalosporins/pharmacology , Escherichia coli/drug effects , Models, Statistical , Penicillanic Acid/analogs & derivatives , Anti-Bacterial Agents/pharmacokinetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cephalosporins/pharmacokinetics , Computer Simulation , Dose-Response Relationship, Drug , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/growth & development , Gene Expression , Microbial Sensitivity Tests , Penicillanic Acid/pharmacokinetics , Penicillanic Acid/pharmacology , Tazobactam , beta-Lactamases/genetics , beta-Lactamases/metabolism
17.
Antimicrob Agents Chemother ; 60(1): 666-8, 2016 01.
Article in English | MEDLINE | ID: mdl-26552971

ABSTRACT

The in vitro activities of ceftolozane-tazobactam, meropenem, and metronidazole were determined against anaerobic organisms isolated from patients with complicated intraabdominal infections (cIAI) in global phase III studies. Ceftolozane-tazobactam activity was highly variable among different species of the Bacteroides fragilis group, with MIC90 values ranging from 2 to 64 µg/ml. More-potent in vitro activity was observed against selected Gram-positive anaerobic organisms; however, small numbers of isolates were available, and, therefore, the clinical significance of these results is unknown. Variable activity of ceftolozane-tazobactam against anaerobic organisms necessitates use in combination with metronidazole for the treatment of cIAI.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacteroides Infections/drug therapy , Cephalosporins/therapeutic use , Gram-Positive Bacterial Infections/drug therapy , Intraabdominal Infections/drug therapy , Metronidazole/therapeutic use , Penicillanic Acid/analogs & derivatives , Anaerobiosis , Bacteroides/drug effects , Bacteroides/isolation & purification , Bacteroides Infections/microbiology , Bacteroides fragilis/drug effects , Bacteroides fragilis/isolation & purification , Drug Therapy, Combination/methods , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/isolation & purification , Gram-Positive Bacterial Infections/microbiology , Humans , Intraabdominal Infections/microbiology , Microbial Sensitivity Tests , Penicillanic Acid/therapeutic use , Tazobactam
18.
Antimicrob Agents Chemother ; 58(10): 6024-31, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25070105

ABSTRACT

It is important to understand the relationship between antibiotic exposure and the selection of drug resistance in the context of therapy exposure. We sought to identify the ceftolozane-tazobactam exposure necessary to prevent the amplification of drug-resistant bacterial subpopulations in a hollow-fiber infection model. Two Pseudomonas aeruginosa challenge isolates were selected for study, a wild-type ATCC strain (ceftolozane-tazobactam MIC, 0.5 mg/liter) and a clinical isolate (ceftolozane-tazobactam MIC, 4 mg/liter). The experiment duration was 10 days, and the ceftolozane-tazobactam dose ratio (2:1) and dosing interval (every 8 h) were selected to approximate those expected to be used clinically. The studied ceftolozane-tazobactam dosing regimens ranged from 62.5/31.25 to 2,000/1,000 mg per dose in step fold dilutions. Negative-control arms included no treatment and tazobactam at 500 mg every 8 h. Positive-control arms included ceftolozane at 1 g every 8 h and piperacillin-tazobactam dosed at 4.5 g every 6 h. For the wild-type ATCC strain, resistance was not selected by any ceftolozane-tazobactam regimen evaluated. For the clinical isolate, an inverted-U-shaped function best described the relationship between the amplification of a drug-resistant subpopulation and drug exposure. The least (62.5/31.25 mg) and most (2,000/1,000 mg) intensive ceftolozane-tazobactam dosing regimens did not select for drug resistance. Drug resistance selection was observed with intermediately intensive dosing regimens (125/62.5 through 1,000/500 mg). For the intermediately intensive ceftolozane-tazobactam dosing regimens, the duration until the selection for drug resistance increased with dose regimen intensity. These data support the selection of ceftolozane-tazobactam dosing regimens that minimize the potential for on-therapy drug resistance selection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Penicillanic Acid/analogs & derivatives , Pseudomonas aeruginosa/drug effects , Microbial Sensitivity Tests , Penicillanic Acid/pharmacology , Tazobactam
19.
Antimicrob Agents Chemother ; 57(12): 5924-30, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24041895

ABSTRACT

We recently investigated the pharmacokinetics-pharmacodynamics (PK-PD) of tazobactam in combination with ceftolozane against an isogenic CTX-M-15-producing Escherichia coli triplet set, genetically engineered to transcribe different levels of blaCTX-M-15. The percentage of the dosing interval that tazobactam concentrations remained above a threshold (%Time>threshold) was identified as the PK-PD exposure measure that was most closely associated with efficacy. Moreover, the tazobactam concentration was dependent upon the enzyme transcription level. Given that the aforementioned strains were genetically engineered to transcribe a single ß-lactamase enzyme and that clinical isolates typically produce multiple ß-lactamase enzymes with various transcription levels, it is likely that the tazobactam threshold concentration is isolate/enzyme dependent. Our first objective was to characterize the relationship between the tazobactam %Time>threshold in combination with ceftolozane and efficacy using clinical isolates in an in vitro PK-PD infection model. Our second objective was to identify a translational relationship that would allow for the comodeling across clinical isolates. The initial challenge panel included four well-characterized ß-lactamase-producing E. coli strains with variable enzyme expression and other resistance determinants. As evidenced by r(2) values of ranging from 0.90 to 0.99 for each clinical isolate, the observed data were well described by fitted functions describing the relationship between the tazobactam %Time>threshold and change in log10 CFU from baseline; however, the data from the four isolates did not comodel well. The threshold concentration identified for each isolate ranged from 0.5 to 4 mg/liter. We identified an enabling translational relationship for the tazobactam threshold that allowed comodeling of all four clinical isolates, which was the product of the individual isolate's ceftolozane-tazobactam MIC value and 0.5. As evidenced by an r(2) value of 0.90, the transformed data were well described by a fitted function describing the relationship between tazobactam %Time>threshold and change in log10 CFU from baseline. Due to these findings, the challenge panel was expanded to include three well-characterized ß-lactamase-producing Klebsiella pneumoniae strains with variable enzyme expression and other resistance determinants. The translational relationship for the tazobactam threshold that allowed for the comodeling of the four E. coli isolates performed well for the expanded data set (seven isolates in total; four E. coli and three K. pneumoniae), as evidenced by an r(2) value of 0.84. This simple translational relationship is especially useful as it is directly linked to in vitro susceptibility test results, which are used to guide the clinician's choice of drug and dosing regimen.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Cephalosporins/pharmacokinetics , Escherichia coli/drug effects , Klebsiella pneumoniae/drug effects , Models, Statistical , Penicillanic Acid/analogs & derivatives , Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Colony Count, Microbial , Computer Simulation , Drug Administration Schedule , Drug Combinations , Drug Dosage Calculations , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Gene Expression , Half-Life , Humans , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology , Klebsiella pneumoniae/enzymology , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/growth & development , Microbial Sensitivity Tests , Penicillanic Acid/pharmacokinetics , Penicillanic Acid/pharmacology , Plasmids , Tazobactam , beta-Lactamases/genetics , beta-Lactamases/metabolism
20.
Antimicrob Agents Chemother ; 57(6): 2809-14, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23629705

ABSTRACT

Despite ß-lactamase inhibitors being available for clinical use for nearly 30 years, a paucity of data exists describing the pharmacokinetic-pharmacodynamic (PK-PD) determinants of efficacy for these agents. Herein, we describe dose fractionation studies designed to determine the exposure measure most predictive of tazobactam efficacy in combination with ceftolozane and the magnitude of this measure necessary for efficacy in a PK-PD in vitro infection model. The challenge organism panel was comprised of an isogenic CTX-M-15-producing Escherichia coli triplet set, genetically engineered to transcribe different levels of bla(CTX-M-15). These recombinant strains exhibited ceftolozane MIC values of 4, 16, and 64 µg/ml representing low, moderate, and high levels of CTX-M-15, respectively. Different bla(CTX-M-15) transcription levels were confirmed by relative quantitative real-time PCR (qRT-PCR) and ß-lactamase hydrolytic assays. The exposure measure associated with efficacy was the percentage of the dosing interval that tazobactam concentrations remained above a threshold (%Time>threshold), regardless of enzyme expression (r(2) = 0.938). The threshold concentrations identified were 0.05 µg/ml for low and moderate and 0.25 µg/ml for the high-ß-lactamase expression strain constructs. The magnitudes of %Time>threshold for tazobactam associated with net bacterial stasis and a 1- and 2-log10 CFU reduction in bacteria at 24 h were approximately 35, 50, and 70%, respectively. These data provide an initial target tazobactam concentration-time profile and a paradigm to optimize tazobactam dosing when combined with ceftolozane.


Subject(s)
Anti-Bacterial Agents , Cephalosporins/administration & dosage , Escherichia coli Infections/drug therapy , Escherichia coli/drug effects , Models, Biological , Penicillanic Acid/analogs & derivatives , beta-Lactamases/biosynthesis , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Area Under Curve , Cephalosporins/pharmacokinetics , Cephalosporins/pharmacology , Cephalosporins/therapeutic use , Dose-Response Relationship, Drug , Drug Therapy, Combination , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Infections/microbiology , Humans , Microbial Sensitivity Tests , Molecular Sequence Data , Penicillanic Acid/administration & dosage , Penicillanic Acid/pharmacokinetics , Penicillanic Acid/pharmacology , Penicillanic Acid/therapeutic use , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Tazobactam , beta-Lactamases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...