Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Antimicrob Chemother ; 79(6): 1353-1361, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38656557

RESUMEN

BACKGROUND: The clinical candidate alpibectir augments the activity of, and overcomes resistance to, the anti-TB drug ethionamide in vitro and in vivo. OBJECTIVES: A Phase 1, double-blind, randomized, placebo-controlled study to investigate the safety, tolerability, pharmacokinetics (PK) and food effect of alpibectir administered as single and multiple oral doses in healthy volunteers (NCT04654143). METHODS: Eighty participants were randomized. In single ascending dose (SAD), a total of six dose levels of alpibectir (0.5 to 40 mg) were tested under fasted and fed (10 mg) conditions as single daily doses in sequential cohorts. In multiple ascending dose (MAD), repeat doses (5 to 30 mg) were administered once daily for 7 days in three sequential cohorts. RESULTS: No serious adverse event was reported. Thirteen participants across groups experienced a total of 13 mild or moderate treatment-emergent adverse events. Alpibectir showed rapid absorption after single dose (mean Tmax range of 0.88 to 1.53 h). Food affected the PK of alpibectir, characterized by a slower absorption (mean Tmax 3.87 h), a lower Cmax (-17.7%) and increased AUC0-t (+19.6%) compared with the fasted condition. Following repeat dosing, dose proportionality was shown for both Cmax and AUC0-tau. Accumulation of alpibectir was observed across all doses, with a more profound effect on AUC during a dosing interval (AUC0-tau) compared with Cmax (1.8- and 1.3-fold on average), respectively. Steady state was considered to have been achieved by Day 7 of dosing. CONCLUSIONS: Alpibectir was generally well tolerated, and no clinically relevant safety findings were identified in the participants treated during SAD or MAD. The PK is dose-proportional and affected by food.


Asunto(s)
Antituberculosos , Voluntarios Sanos , Humanos , Adulto , Masculino , Femenino , Método Doble Ciego , Adulto Joven , Persona de Mediana Edad , Antituberculosos/farmacocinética , Antituberculosos/administración & dosificación , Antituberculosos/efectos adversos , Interacciones Alimento-Droga , Administración Oral , Adolescente , Placebos/administración & dosificación , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos
2.
Eur J Med Chem ; 238: 114515, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-35691174

RESUMEN

Acinetobacter baumannii is a gram-negative bacterium causing severe hospital-acquired infections such as bloodstream infections or pneumonia. Moreover, multidrug resistant A. baumannii becomes prevalent in many hospitals. Consequently, the World Health Organization made this bacterium a critical priority for the research and development of new antibiotics. Rifabutin, a semisynthetic product from the rifamycin class, was recently found to be very active in nutrient-limited eukaryotic cell culture medium against various A. baumannii strains, including extremely drug-resistant strains, with minimal inhibitory concentrations as low as 0.008 µg/mL. Moreover, this in vitro potency translates into in vivo efficacy. Thus, rifabutin appears to be an attractive novel antibiotic against A. baumannii. In this work, our objective was to design and synthetize rifabutin prodrugs with increased aqueous solubility to allow intraveneous use. Synthetic methodologies were developed to selectively functionalize the hydroxyl group in position 21 and to afford 17 prodrugs. We measured the water solubility of the prodrugs, the stability in human and mouse plasma and their antimicrobial activity against A. baumannii after incubation in human serum. Finally, a pharmacokinetic release study of rifabutin was performed in CD1 mice with three selected prodrugs as a proof of concept.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Profármacos , Infecciones por Acinetobacter/tratamiento farmacológico , Infecciones por Acinetobacter/microbiología , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana Múltiple , Ratones , Pruebas de Sensibilidad Microbiana , Profármacos/farmacología , Rifabutina/farmacología , Agua
3.
Drug Discov Today ; 26(9): 2099-2104, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34242796

RESUMEN

Rifamycin antibiotics were discovered during the 1950s, and their main representative, rifampicin, remains a cornerstone treatment for TB. The clinical use of rifamycin is restricted to mycobacteria and Gram-positive infections because of its poor ability to penetrate the Gram-negative outer membrane. Rifabutin, a rifamycin antibiotic approved for the prevention of Mycobacterium avium complex disease, makes an exception to this rule by hijacking the iron uptake system of Acinetobacter baumannii, resulting in potent activity against this important Gram-negative pathogen. Here, we describe recent findings on the specific activity of rifabutin and provide evidence of the need for the development of an intravenous formulation of rifabutin (BV100) for the treatment of difficult-to-treat carbapenem-resistant A.baumannii infections.


Asunto(s)
Infecciones por Acinetobacter/tratamiento farmacológico , Antibacterianos/administración & dosificación , Rifabutina/administración & dosificación , Acinetobacter baumannii/efectos de los fármacos , Animales , Antibacterianos/farmacocinética , Carbapenémicos , Farmacorresistencia Bacteriana , Humanos , Infusiones Intravenosas , Rifabutina/farmacocinética
4.
J Antimicrob Chemother ; 75(12): 3552-3562, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32869081

RESUMEN

BACKGROUND: Rifabutin, an oral drug approved to treat Mycobacterium avium infections, demonstrated potent activity against Acinetobacter baumannii in nutrient-limited medium enabled by rifabutin cellular uptake through the siderophore receptor FhuE. OBJECTIVES: To determine rifabutin in vitro activity and resistance mechanisms in a large panel of A. baumannii isolates. METHODS: Two hundred and ninety-three carbapenem-resistant A. baumannii clinical isolates collected from Europe, the USA and Asia during 2017-19 were used for MIC determination. Sequencing/genotyping of fhuE, rpoB and arr-2 genes in isolates with elevated rifabutin MIC combined with genetic engineering and gene expression quantification was used to characterize rifabutin's mode of action and resistance mechanisms. RESULTS: Rifabutin showed excellent activity on the strain panel, with an MIC50/90 of 0.008/1 mg/L, and was superior to all other antibiotics tested, including colistin, tigecycline and cefiderocol (MIC90 of 8 mg/L). Rifabutin remained active on resistant subpopulations, including strains resistant to the siderophore-drug conjugate cefiderocol (MIC90 of 2 mg/L, n = 23). At least two independent resistance mechanisms were required to abolish rifabutin activity, which is in line with the dose-dependent mutational resistance frequency reaching 10-9 at rifabutin concentrations at or above 2 mg/L. CONCLUSIONS: This study demonstrated the potent activity of rifabutin against carbapenem-resistant A. baumannii. We propose that FhuE-mediated active uptake of rifabutin enables activity against rifampicin-resistant isolates. To achieve clinically meaningful strain coverage and to avoid rapid resistance development, rifabutin concentrations ≥2 mg/L are required, something rifabutin oral formulations cannot deliver.


Asunto(s)
Acinetobacter baumannii , Acinetobacter baumannii/genética , Antibacterianos/farmacología , Asia , Carbapenémicos/farmacología , Farmacorresistencia Bacteriana Múltiple , Europa (Continente) , Pruebas de Sensibilidad Microbiana , Rifabutina/farmacología
5.
Nat Microbiol ; 5(9): 1134-1143, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32514072

RESUMEN

Industry screens of large chemical libraries have traditionally relied on rich media to ensure rapid bacterial growth in high-throughput testing. We used eukaryotic, nutrient-limited growth media in a compound screen that unmasked a previously unknown hyperactivity of the old antibiotic, rifabutin (RBT), against highly resistant Acinetobacter baumannii. In nutrient-limited, but not rich, media, RBT was 200-fold more potent than rifampin. RBT was also substantially more effective in vivo. The mechanism of enhanced efficacy was a Trojan horse-like import of RBT, but not rifampin, through fhuE, only in nutrient-limited conditions. These results are of fundamental importance to efforts to discover antibacterial agents.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Nutrientes/metabolismo , Rifabutina/farmacología , Infecciones por Acinetobacter/tratamiento farmacológico , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/genética , Animales , Proteínas Bacterianas/efectos de los fármacos , Proteínas Bacterianas/genética , Colistina/farmacología , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple/genética , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Masculino , Ratones , Ratones Endogámicos C3H , Pruebas de Sensibilidad Microbiana , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/genética , Rifampin/farmacología
6.
J Antimicrob Chemother ; 75(6): 1518-1524, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32087024

RESUMEN

BACKGROUND: POL7306 belongs to a new class of peptidomimetic outer-membrane-protein-targeting antibiotics with a novel mechanism of action. POL7306 is in development for the treatment of infections caused by antimicrobial-resistant Gram-negative bacteria and has demonstrated low cytotoxicity and nephrotoxicity. METHODS: A total of 891 isolates were collected by the SENTRY Antimicrobial Surveillance Program from 134 medical centres in Europe (n = 424; 41 centres in 18 nations), the USA (n = 411 isolates from 67 centres), the Asia-Pacific region (n = 24; 15 centres in 6 nations) and Latin America (n = 32; 11 centres in 9 nations) and included 558 Enterobacterales, 310 non-fermenters and 23 fastidious organisms. Susceptibility testing was performed using the reference broth microdilution method and the medium was supplemented with 0.002% polysorbate-80 for testing POL7306. Resistant subsets were characterized by WGS. RESULTS: POL7306 demonstrated potent in vitro activity against Enterobacterales [including carbapenem-resistant (MIC50/90, 0.06/0.25 mg/L), ESBL-producing (MIC50/90, 0.06/0.12 mg/L), KPC-producing (MIC50/90, 0.12/0.25 mg/L), MBL-producing (MIC50/90, 0.06/0.25 mg/L), colistin-non-susceptible, mcr-negative (MIC50/90, 0.5/2 mg/L) and mcr-positive (MIC50/90, 0.12/0.25 mg/L) Enterobacterales], Pseudomonas aeruginosa (MIC50/90, 0.25/0.25 mg/L), Acinetobacter baumannii (MIC50/90, 0.06/0.12 mg/L) and Stenotrophomonas maltophilia (MIC50/90, 0.06/0.25 mg/L). CONCLUSIONS: POL7306 demonstrated potent activity against a large collection of Gram-negative organisms collected worldwide that included colistin-resistant, XDR and ESBL- and carbapenemase-producing isolates for which there are currently limited treatment options.


Asunto(s)
Farmacorresistencia Bacteriana Múltiple , Bacterias Gramnegativas , Antibacterianos/farmacología , Asia , Europa (Continente) , América Latina , Pruebas de Sensibilidad Microbiana
7.
Artículo en Inglés | MEDLINE | ID: mdl-31767727

RESUMEN

The objective was to determine the in vitro antimicrobial susceptibility of Pseudomonas aeruginosa isolates cultured from cystic fibrosis (CF) patients and explore associations between strain sequence type and susceptibility. Fourteen antibiotics and antibiotic combinations, including the novel antibacterial peptide murepavadin, were tested for activity against 414 Pseudomonas aeruginosa isolates cultured from respiratory samples of CF patients. The complete genomes of the isolates were sequenced, and minimum spanning trees were constructed based on the sequence types (STs). Percentages of resistance according to CLSI 2019 breakpoints were as follows: cefepime, 14%; ceftazidime, 11%; ceftazidime-avibactam, 7%; ceftolozane-tazobactam, 3%; piperacillin-tazobactam, 12%; meropenem, 18%; imipenem, 32%; aztreonam, 23%; ciprofloxacin, 30%; gentamicin, 30%; tobramycin, 12%; amikacin, 18%; and colistin, 4%. Murepavadin MIC50 and MIC90 were 0.12 mg/liter and 2 mg/liter, respectively. There were no apparent clonal clusters associated with resistance, but higher MICs did appear to occur more often in STs with multiple isolates than in single ST isolates. In general, the CF isolates showed a wide genetic distribution. P. aeruginosa CF isolates exhibited the lowest resistance rates against ceftolozane-tazobactam, ceftazidime-avibactam, and colistin. Murepavadin demonstrated the highest activity on a per-weight basis and may therefore become a valuable addition to the currently available antibiotics for treatment of respiratory infection in people with CF.


Asunto(s)
Antibacterianos/farmacología , Fibrosis Quística/microbiología , Péptidos Cíclicos/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Farmacorresistencia Bacteriana Múltiple , Humanos , Pruebas de Sensibilidad Microbiana , Tipificación de Secuencias Multilocus , Infecciones por Pseudomonas , Pseudomonas aeruginosa/genética
9.
Nature ; 576(7787): 452-458, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31645764

RESUMEN

There is an urgent need for new antibiotics against Gram-negative pathogens that are resistant to carbapenem and third-generation cephalosporins, against which antibiotics of last resort have lost most of their efficacy. Here we describe a class of synthetic antibiotics inspired by scaffolds derived from natural products. These chimeric antibiotics contain a ß-hairpin peptide macrocycle linked to the macrocycle found in the polymyxin and colistin family of natural products. They are bactericidal and have a mechanism of action that involves binding to both lipopolysaccharide and the main component (BamA) of the ß-barrel folding complex (BAM) that is required for the folding and insertion of ß-barrel proteins into the outer membrane of Gram-negative bacteria. Extensively optimized derivatives show potent activity against multidrug-resistant pathogens, including all of the Gram-negative members of the ESKAPE pathogens1. These derivatives also show favourable drug properties and overcome colistin resistance, both in vitro and in vivo. The lead candidate is currently in preclinical toxicology studies that-if successful-will allow progress into clinical studies that have the potential to address life-threatening infections by the Gram-negative pathogens, and thus to resolve a considerable unmet medical need.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Farmacorresistencia Microbiana , Bacterias Gramnegativas/efectos de los fármacos , Peptidomiméticos/química , Peptidomiméticos/farmacología , Animales , Antibacterianos/efectos adversos , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Productos Biológicos/química , Descubrimiento de Drogas , Farmacorresistencia Microbiana/efectos de los fármacos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Fluorescencia , Bacterias Gramnegativas/genética , Bacterias Gramnegativas/patogenicidad , Humanos , Lipopolisacáridos/química , Compuestos Macrocíclicos/efectos adversos , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacología , Masculino , Ratones , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Microscopía Electrónica de Transmisión , Modelos Moleculares , Mutación , Peptidomiméticos/efectos adversos , Etiquetas de Fotoafinidad
10.
Artículo en Inglés | MEDLINE | ID: mdl-30012756

RESUMEN

This open-label, nonrandomized, single-dose, phase 1 study evaluated the pharmacokinetics and safety of murepavadin, a novel peptide antibiotic for the treatment of serious Pseudomonas aeruginosa infections. The study was conducted in 32 subjects of either sex in 4 groups (up to 8 per group) with mild (group 1), moderate (group 2), and severe (group 3) renal function impairment or with normal renal function (group 4). The degree of renal impairment of the subjects was classified at screening according to the estimated creatinine clearance (CLCr) according to the Cockcroft-Gault equation. All subjects received a single 2.2-mg/kg of body weight intravenous infusion of murepavadin administered over 3 h. Exposure to murepavadin in plasma increased in subjects with renal function impairment, with the area under the plasma concentration-time curve from zero to infinity (AUC0-∞) increasing about 2.0- to 2.5-fold for subjects with renal function impairment compared to subjects with normal renal function, whereas the increases in maximum observed plasma concentration (Cmax) were about 1.5-fold for subjects with renal function impairment compared to subjects with normal renal function. The total clearance (CL) of murepavadin was lower in all groups of subjects with renal function impairment, with group means ranging from 2.4 liters/h to 3.8 liters/h, compared to 7.0 liters/h in subjects with normal renal function. Accordingly, the terminal elimination half-life (t1/2) prolonged up to 24 h with decreasing renal function compared to 7.7 h in subjects with normal renal function. Murepavadin was well tolerated in all renal function groups. As the elimination of murepavadin is affected by renal function, a dose adjustment is warranted in subjects with impaired renal function. (This paper has been registered at ClinicalTrials.gov under identifier NCT02110459.).


Asunto(s)
Antibacterianos/efectos adversos , Antibacterianos/farmacocinética , Péptidos Cíclicos/efectos adversos , Péptidos Cíclicos/farmacocinética , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Insuficiencia Renal/metabolismo , Adulto , Anciano , Área Bajo la Curva , Femenino , Humanos , Riñón/metabolismo , Masculino , Persona de Mediana Edad , Infecciones por Pseudomonas/metabolismo
11.
J Antimicrob Chemother ; 73(9): 2400-2404, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29901750

RESUMEN

Background: Murepavadin (POL7080) represents the first member of a novel class of outer membrane protein-targeting antibiotics. Murepavadin acts by binding to LPS transport protein D and is being developed for the treatment of hospital-acquired and ventilator-associated pneumonia caused by Pseudomonas aeruginosa. Objectives: To evaluate the antimicrobial activity of murepavadin against XDR P. aeruginosa. Methods: A total of 785 clinical isolates of XDR P. aeruginosa were collected in 2016-17 through the SENTRY Antimicrobial Surveillance Program from 34 medical centres in 21 European nations (n = 353) and 75 medical centres in North America (n = 432). Isolates were categorized as XDR when susceptible (CLSI) to ≤2 of the following antimicrobial classes: antipseudomonal cephalosporins, carbapenems, broad-spectrum penicillin/ß-lactamase inhibitor combinations, fluoroquinolones, aminoglycosides and polymyxins. Susceptibility testing was performed by the reference broth microdilution method and EUCAST and CLSI interpretative criteria were applied. Results: Murepavadin (MIC50/90, 0.12/0.25 mg/L) inhibited 96.7% of isolates at ≤0.5 mg/L and was 8-fold more active than colistin (MIC50/90, 1/2 mg/L). Only seven isolates (0.9%) exhibited murepavadin MIC values >4 mg/L. Colistin (MIC50/90, 1/2 mg/L; 93.6% susceptible) was the most active comparator, followed by ceftolozane/tazobactam (MIC50/90, 2/>32 mg/L; 70.6% susceptible) and tobramycin (MIC50/90, 8/>8 mg/L; 47.5% susceptible). Murepavadin remained active against isolates that were non-susceptible to colistin (n = 50; MIC50/90, 0.25/0.25 mg/L), ceftolozane/tazobactam (n = 231; MIC50/90, 0.12/0.25 mg/L) and/or tobramycin (n = 412; MIC50/90, 0.12/0.25 mg/L). Conclusions: Murepavadin exhibited potent activity against a large collection of clinical XDR P. aeruginosa isolates from Europe and North America, including isolates that were non-susceptible to colistin, ceftolozane/tazobactam and/or tobramycin.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple , Péptidos Cíclicos/farmacología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Aspartato Aminotransferasas/sangre , Europa (Continente) , Humanos , Pruebas de Sensibilidad Microbiana , América del Norte , Pseudomonas aeruginosa/aislamiento & purificación
12.
Artículo en Inglés | MEDLINE | ID: mdl-29686157

RESUMEN

Murepavadin (formerly POL7080), a 14-amino-acid cyclic peptide, and comparators were tested by the broth microdilution method against 1,219 Pseudomonas aeruginosa isolates from 112 medical centers. Murepavadin (MIC50/90, 0.12/0.12 mg/liter) was 4- to 8-fold more active than colistin (MIC50/90, 1/1 mg/liter) and polymyxin B (MIC50/90, 0.5/1 mg/liter) and inhibited 99.1% of isolates at ≤0.5 mg/liter. Only 4 isolates (0.3%) exhibited murepavadin MICs of >2 mg/liter. Murepavadin was equally active against isolates from Europe, the United States, and China.


Asunto(s)
Antibacterianos/farmacología , Péptidos Cíclicos/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , China , Colistina/farmacología , Farmacorresistencia Bacteriana Múltiple , Europa (Continente) , Humanos , Pruebas de Sensibilidad Microbiana , Polimixina B/farmacología , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/aislamiento & purificación , Estados Unidos
13.
Artículo en Inglés | MEDLINE | ID: mdl-29437621

RESUMEN

Murepavadin is the first in class of the outer membrane protein-targeting antibiotics (OMPTA) and a pathogen-specific peptidomimetic antibacterial with a novel, nonlytic mechanism of action targeting Pseudomonas aeruginosa Murepavadin is being developed for the treatment of hospital-acquired bacterial pneumonia (HABP) and ventilator-associated bacterial pneumonia (VABP). The pharmacokinetics (PK) and safety of single and multiple doses of murepavadin were investigated in healthy male subjects. Part A of the study was a double-blind, randomized, placebo-controlled, single-ascending-dose investigation in 10 sequential cohorts where each cohort comprised 6 healthy male subjects; 4 subjects were randomized to murepavadin, and 2 subjects were randomized to placebo. Part B was a double-blind, randomized, placebo-controlled, multiple-ascending-dose investigation in 3 sequential cohorts. After a single dose of murepavadin, the geometric mean half-life (2.52 to 5.30 h), the total clearance (80.1 to 114 ml/h/kg), and the volume of distribution (415 to 724 ml/kg) were consistent across dose levels. The pharmacokinetics of the dosing regimens evaluated were dose proportional and linear. Murepavadin was well tolerated, adverse events were transient and generally mild, and no dose-limiting toxicity was identified.


Asunto(s)
Antibacterianos/efectos adversos , Antibacterianos/farmacocinética , Péptidos Cíclicos/efectos adversos , Péptidos Cíclicos/farmacocinética , Adulto , Antibacterianos/administración & dosificación , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Voluntarios Sanos , Humanos , Infusiones Intravenosas , Masculino , Péptidos Cíclicos/administración & dosificación , Adulto Joven
14.
Expert Rev Anti Infect Ther ; 16(4): 259-268, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29451043

RESUMEN

INTRODUCTION: With the increase in multi-drug resistant P. aeruginosa, antimicrobials with a novel mechanism of action are needed that can target these infections. Areas covered: Intravenous murepavadin is in Phase 3 development for the treatment of HABP/VABP due to P. aeruginosa. This paper summarizes the available information on the discovery, the in vitro activity, pharmacokinetic and pharmacodynamic properties and the clinical pharmacology of murepavadin to date. Expert commentary: P. aeruginosa has an intrinsic resistance to many antibiotics due to high cellular impermeability and efficient drug efflux mechanisms, and the recent increase in prevalence of multidrug-resistant P. aeruginosa infections are particularly threatening in ICU settings. Murepavadin is a pathogen specific antimicrobial peptidomimetic with a novel, non-lytic mechanism of action, and is the first in class of outer membrane protein targeting antibiotics which are being developed. Murepavadin displays a potent in vitro activity including carbapenemase-producing and colistin-resistant P. aeruginosa. Murepavadin is active in pre-clinical animal models including infections with XDR isolates. The Pharmacokinetics is well characterized including subjects with impaired renal function and patients with VABP. Intravenous murepavadin is currently under clinical development for the treatment of HABP/VABP due to P. aeruginosa infection.


Asunto(s)
Antibacterianos/farmacología , Péptidos Cíclicos/farmacología , Infecciones por Pseudomonas/tratamiento farmacológico , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacocinética , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Humanos , Péptidos Cíclicos/administración & dosificación , Péptidos Cíclicos/farmacocinética , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/aislamiento & purificación
15.
Chest ; 152(2): 249-262, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28442313

RESUMEN

In many respiratory diseases characterized by an intense inflammatory response, the balance between proteolytic enzymes (proteases, including elastases) and their inhibitors (proteinases inhibitors) is not neutral. Excess activity of neutrophil elastase (NE) and similar proteases has been reported to cause tissue damage and to alter the remodeling process in many clinical conditions such as pneumonia, respiratory distress, and acute lung injury (ALI). Several experimental NE inhibitors have been tested in preclinical and clinical studies of different conditions of inflammatory lung injury such as ALI and pneumonia, with contrasting results. This study reviews the literature regarding NE inhibitors in the field of respiratory diseases and reflects on possible future developments. In particular, we highlight potential gaps in the scientific evidence and discuss potential strategies for focusing investigation on antielastases in clinical practice through the selection of targeted populations and proper outcomes.


Asunto(s)
Enfermedades Pulmonares/tratamiento farmacológico , Proteínas Inhibidoras de Proteinasas Secretoras/farmacología , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/enzimología , Animales , Bronquiectasia/tratamiento farmacológico , Bronquiectasia/enzimología , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/enzimología , Modelos Animales de Enfermedad , Humanos , Elastasa de Leucocito/fisiología , Enfermedades Pulmonares/enzimología , Proteínas Inhibidoras de Proteinasas Secretoras/fisiología , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/enzimología
16.
Antimicrob Agents Chemother ; 60(8): 4991-5000, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27297477

RESUMEN

The clinical development of antibiotics with a new mode of action combined with efficient pulmonary drug delivery is a priority against untreatable Pseudomonas aeruginosa lung infections. POL7001 is a macrocycle antibiotic belonging to the novel class of protein epitope mimetic (PEM) molecules with selective and potent activity against P. aeruginosa We investigated ventilator-associated pneumonia (VAP) and cystic fibrosis (CF) as indications of the clinical potential of POL7001 to treat P. aeruginosa pulmonary infections. MICs of POL7001 and comparators were measured for reference and clinical P. aeruginosa strains. The therapeutic efficacy of POL7001 given by pulmonary administration was evaluated in murine models of P. aeruginosa acute and chronic pneumonia. POL7001 showed potent in vitro activity against a large panel of P. aeruginosa isolates from CF patients, including multidrug-resistant (MDR) isolates with adaptive phenotypes such as mucoid or hypermutable phenotypes. The efficacy of POL7001 was demonstrated in both wild-type and CF mice. In addition to a reduced bacterial burden in the lung, POL7001-treated mice showed progressive body weight recovery and reduced levels of inflammatory markers, indicating an improvement in general condition. Pharmacokinetic studies indicated that POL7001 reached significant concentrations in the lung after pulmonary administration, with low systemic exposure. These results support the further evaluation of POL7001 as a novel therapeutic agent for the treatment of P. aeruginosa pulmonary infections.


Asunto(s)
Antibacterianos/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Animales , Antibacterianos/farmacocinética , Fibrosis Quística/microbiología , Pulmón/efectos de los fármacos , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Neumonía Asociada al Ventilador/microbiología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Infecciones del Sistema Respiratorio/microbiología
17.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 8): 751-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19622858

RESUMEN

Iclaprim is a novel dihydrofolate reductase (DHFR) inhibitor belonging to the 2,4-diaminopyrimidine class of antibiotics, of which trimethoprim (TMP) is the most well known representative. Iclaprim exhibits potent bactericidal activity against major Gram-positive pathogens, notably methicillin-sensitive Staphylococcus aureus (MSSA) and methicillin-resistant S. aureus (MRSA) phenotypes, including TMP-resistant strains. The inhibition properties of racemic iclaprim and of the two enantiomers, termed AR-101 and AR-102, towards S. aureus wild-type DHFR and TMP-resistant F98Y mutant DHFR were determined and compared. Similar to TMP, AR-101, AR-102 and iclaprim are all competitive inhibitors with respect to the substrate dihydrofolate. Iclaprim, AR-101 and AR-102 demonstrated little or no difference in activity towards these enzymes and were significantly more potent than TMP. The crystal structures of S. aureus DHFR and F98Y mutant DHFR were determined as ternary complexes with NADPH and either AR-101, AR-102 or iclaprim. The binding modes of the inhibitors were analysed and compared. The X-ray crystallographic data explain the binding modes of all molecules well and can be used to rationalize the equipotent affinity of AR-101, AR-102 and iclaprim, which is also reflected in their antibacterial properties.


Asunto(s)
Antibacterianos/química , Inhibidores Enzimáticos/química , Staphylococcus aureus Resistente a Meticilina/enzimología , Proteínas Mutantes/metabolismo , Pirimidinas/química , Tetrahidrofolato Deshidrogenasa/metabolismo , Trimetoprim/química , Antibacterianos/metabolismo , Cristalización , Cristalografía por Rayos X , Inhibidores Enzimáticos/metabolismo , Isomerismo , Resistencia a la Meticilina , Complejos Multienzimáticos/química , Complejos Multienzimáticos/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/genética , NADP/química , NADP/metabolismo , Estructura Terciaria de Proteína , Pirimidinas/metabolismo , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/genética , Trimetoprim/metabolismo
18.
J Antimicrob Chemother ; 63(4): 687-98, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19211577

RESUMEN

OBJECTIVES: Iclaprim is a novel 2,4-diaminopyrimidine that exhibits potent, rapid bactericidal activity against major Gram-positive pathogens, including methicillin-susceptible Staphylococcus aureus and methicillin-resistant S. aureus, and is currently in clinical development for the treatment of complicated skin and skin structure infections. An understanding of the known mechanism of resistance to trimethoprim led to the design of this new inhibitor, with improved affinity towards dihydrofolate reductase (DHFR) from S. aureus and clinically useful activity against S. aureus including isolates resistant to trimethoprim. The objective of this study was to characterize the mode of action of iclaprim and its inhibitory properties against DHFR. METHODS: The mode of action of iclaprim was assessed by enzymatic analysis, direct binding studies, macromolecular synthesis profiles, synergy and antagonism studies to define its role as an inhibitor of DHFR. The binding properties of iclaprim to DHFR were compared with those of trimethoprim by X-ray crystallography. RESULTS: The enzymatic properties, direct binding and X-ray crystallographic studies delineated the mode of interaction with DHFR and the reason for the increased affinity of iclaprim towards the enzyme. The effect of iclaprim on bacterial physiology suggests that iclaprim behaves as a classical antibacterial DHFR inhibitor, as previously documented for trimethoprim. CONCLUSIONS: Iclaprim binds and inhibits bacterial DHFR in a similar manner to trimethoprim. However, the increased hydrophobic interactions between iclaprim and DHFR account for increased affinity and, unlike trimethoprim, enable iclaprim to inhibit even the resistant enzyme with nanomolar affinity, thus overcoming the mechanism of trimethoprim resistance. The increased antibacterial activity and lower propensity for resistance make iclaprim a clinically promising and useful inhibitor.


Asunto(s)
Antibacterianos/farmacología , Inhibidores Enzimáticos/farmacología , Pirimidinas/farmacología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/enzimología , Tetrahidrofolato Deshidrogenasa/metabolismo , Antibacterianos/metabolismo , Cristalografía por Rayos X , Inhibidores Enzimáticos/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Unión Proteica , Estructura Terciaria de Proteína , Pirimidinas/metabolismo , Trimetoprim/metabolismo , Trimetoprim/farmacología
19.
J Mol Biol ; 385(1): 178-87, 2009 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-18992253

RESUMEN

Endothelin-converting enzyme I (ECE-1) is a mammalian type II integral membrane zinc-containing endopeptidase. ECE-1 catalyzes the final step in the biosynthesis of endothelins in a rate-limiting fashion, through post-translational conversion of the biologically inactive big endothelins. Endothelin-1 overproduction has been implicated in a heterogeneous list of diseases including systemic and pulmonary hypertension, stroke and asthma, cardiac and renal failure. Therefore, ECE-1 is a prime therapeutic target for the regulation of endothelin-1 production in vivo and there is considerable interest in selective inhibitors of this enzyme. Here, we present the crystal structure of the extracellular domain (residues 90-770) of human ECE-1 (C428S) with the generic metalloprotease inhibitor phosphoramidon determined at 2.38 A resolution. The structure is closely related to that of human NEP, providing essential information for a detailed understanding of ligand-binding, specificity determinants as well as selectivity criteria. Selective inhibitors of ECE-1s should have beneficial effects for the treatment of diseases in which an overproduction of ETs plays a pathogenic role.


Asunto(s)
Ácido Aspártico Endopeptidasas/química , Glicopéptidos/química , Metaloendopeptidasas/química , Secuencia de Aminoácidos , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Enzimas Convertidoras de Endotelina , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Enlace de Hidrógeno/efectos de los fármacos , Metaloendopeptidasas/antagonistas & inhibidores , Metaloendopeptidasas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Neprilisina/química , Estructura Secundaria de Proteína , Alineación de Secuencia , Especificidad por Sustrato/efectos de los fármacos , Propiedades de Superficie/efectos de los fármacos , Zinc/metabolismo
20.
Acta Crystallogr D Biol Crystallogr ; 63(Pt 9): 975-81, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17704566

RESUMEN

Neutral endopeptidase (NEP) is the major enzyme involved in the metabolic inactivation of a number of bioactive peptides including the enkephalins, substance P, endothelin, bradykinin and atrial natriuretic factor, as well as the incretin hormone glucagon-like peptide 1 (GLP-1), which is a potent stimulator of insulin secretion. The activity of GLP-1 is also rapidly abolished by the serine protease dipeptidyl peptidase IV (DPP-IV), which led to an elevated interest in inhibitors of this enzyme for the treatment of type II diabetes. A dual NEP/DPP-IV inhibitor concept is proposed, offering an alternative strategy for the treatment of type 2 diabetes. Here, the synthesis and crystal structures of the soluble extracellular domain of human NEP (residues 52-749) complexed with the NEP, competitive and potent dual NEP/DPP-IV inhibitor MCB3937 are described.


Asunto(s)
Inhibidores de la Adenosina Desaminasa , Inhibidores de la Dipeptidil-Peptidasa IV , Glicoproteínas/antagonistas & inhibidores , Neprilisina/antagonistas & inhibidores , Neprilisina/metabolismo , Oligopéptidos/química , Oligopéptidos/farmacología , Adenosina Desaminasa/metabolismo , Unión Competitiva , Cristalografía por Rayos X , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dipeptidil Peptidasa 4/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Glicoproteínas/metabolismo , Humanos , Modelos Moleculares , Estructura Molecular , Neprilisina/química , Fragmentos de Péptidos/química , Unión Proteica , Estructura Terciaria de Proteína , Zinc/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...