Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmaceutics ; 14(9)2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36145568

ABSTRACT

The encapsulation of peptides and proteins in nanosystems has been extensively investigated for masking unfavorable biopharmaceutical properties, including short half-life and poor permeation through biological membranes. Therefore, the aim of this work was to encapsulate a small antimicrobial hydrophilic peptide (H-Ser-Pro-Trp-Thr-NH2, FS10) in PEG-PLGA (polyethylene glycol-poly lactic acid-co-glycolic acid) nanoparticles (Nps) and thereby overcome the common limitations of hydrophilic drugs, which because they facilitate water absorption suffer from rapid degradation. FS10 is structurally related to the well-known RNAIII inhibiting peptide (RIP) and inhibits S. aureus biofilm formation. Various parameters, including different method (double emulsion and nanoprecipitation), pH of the aqueous phase and polymeric composition, were investigated to load FS10 into PEG-PLGA nanoparticles. The combination of different strategies resulted in an encapsulation efficiency of around 25% for both the double emulsion and the nanoprecipitation method. It was found that the most influential parameters were the pH­which tailors the peptides charge­and the polymeric composition. FS10-PEG-PLGA nanoparticles, obtained under optimized parameters, showed size lower than 180 nm with zeta potential values ranging from −11 to −21 mV. In vitro release studies showed that the Nps had an initial burst release of 48−63%, followed by a continuous drug release up to 21 h, probably caused by the porous character of the Nps. Furthermore, transmission electron microscopy (TEM) analysis revealed particles with a spherical morphology and size of around 100 nm. Antimicrobial assay showed that the minimum inhibitory concentration (MIC) of the FS10-loaded Nps, against S. aureus strains, was lower (>128 µg/mL) than that of the free FS10 (>256 µg/mL). The main goal of this work was to develop polymeric drug delivery systems aiming at protecting the peptide from a fast degradation, thus improving its accumulation in the target site and increasing the drug-bacterial membrane interactions.

2.
Article in English | MEDLINE | ID: mdl-32540976

ABSTRACT

Antimicrobial peptides (AMPs) have seen limited clinical use as antimicrobial agents, largely due to issues relating to toxicity, short biological half-life, and lack of efficacy against Gram-negative bacteria. However, the development of novel AMP-nanomedicines, i.e., AMPs entrapped in nanoparticles, has the potential to ameliorate these clinical problems. The authors investigated two novel nanomedicines based on AA139, an AMP currently in development for the treatment of multidrug-resistant Gram-negative infections. AA139 was entrapped in polymeric nanoparticles (PNPs) or lipid-core micelles (MCLs). The antimicrobial activity of AA139-PNP and AA139-MCL was determined in vitro The biodistribution and limiting doses of AA139-nanomedicines were determined in uninfected rats via endotracheal aerosolization. The early bacterial killing activity of the AA139-nanomedicines in infected lungs was assessed in a rat model of pneumonia-septicemia caused by extended-spectrum ß-lactamase-producing Klebsiella pneumoniae In this model, the therapeutic efficacy was determined by once-daily (q24h) administration over 10 days. Both AA139-nanomedicines showed equivalent in vitro antimicrobial activities (similar to free AA139). In uninfected rats, they exhibited longer residence times in the lungs than free AA139 (∼20% longer for AA139-PNP and ∼80% longer for AA139-MCL), as well as reduced toxicity, enabling a higher limiting dose. In rats with pneumonia-septicemia, both AA139-nanomedicines showed significantly improved therapeutic efficacy in terms of an extended rat survival time, although survival of all rats was not achieved. These results demonstrate potential advantages that can be achieved using AMP-nanomedicines. AA139-PNP and AA139-MCL may be promising novel therapeutic agents for the treatment of patients suffering from multidrug-resistant Gram-negative pneumonia-septicemia.


Subject(s)
Bacteremia , Drug Resistance, Multiple, Bacterial , Klebsiella Infections/drug therapy , Pneumonia, Bacterial , Pore Forming Cytotoxic Proteins , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteremia/drug therapy , Klebsiella pneumoniae , Microbial Sensitivity Tests , Nanomedicine , Pneumonia, Bacterial/drug therapy , Pore Forming Cytotoxic Proteins/pharmacology , Rats , Tissue Distribution
3.
Future Microbiol ; 13: 1683-1692, 2018 11.
Article in English | MEDLINE | ID: mdl-30499686

ABSTRACT

Respiratory tract infections are one of the most frequent infections worldwide, with an increasing number being associated with (multiple) antibiotic-resistant pathogens. Improved treatment requires the development of new therapeutic strategies, including the possible development of antibiotic-nanomedicines. Antibiotic-nanomedicines comprise antibiotic molecules coupled to nanocarriers via surface adsorption, surface attachment, entrapment or conjugation and can be administered via aerosolization. The efficacy and tolerability of this approach has been shown in clinical studies, with amikacin liposome inhalation suspension being the first inhalatory antibiotic-nanomedicine approved by the US FDA. In this special report, we summarize and discuss the potential value and the clinical status of antibiotic-nanomedicines for the treatment of (antibiotic-resistant) respiratory tract infections.


Subject(s)
Administration, Inhalation , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Nanomedicine/methods , Respiratory Tract Infections/drug therapy , Amikacin/pharmacology , Drug Approval , Drug Resistance, Microbial , Humans , Liposomes , Particle Size , Respiratory System , Respiratory Tract Infections/microbiology , Treatment Outcome , United States , United States Food and Drug Administration
4.
Biomaterials ; 119: 68-77, 2017 03.
Article in English | MEDLINE | ID: mdl-28002754

ABSTRACT

Approximately a dozen of intravenous iron nanomedicines gained marketing authorization in the last two decades. These products are generally considered as safe, but have been associated with an increased risk for hypersensitivity-like reactions of which the underlying mechanisms are unknown. We hypothesized that iron nanomedicines can trigger the innate immune system. We hereto investigated the physico-chemical properties of ferric gluconate, iron sucrose, ferric carboxymaltose and iron isomaltoside 1000 and comparatively studied their interaction with Toll-like receptors, the complement system and peripheral blood mononuclear cells. Two out of four formulations appeared as aggregates by Scanning Transmission Electron Microscopy analysis and were actively taken up by HEK293T- and peripheral blood mononuclear cells in a cholesterol-dependent manner. These formulations triggered in vitro activation of intracellular Toll-like receptors 3, -7 and -9 in a dose- and serum-dependent manner. In parallel experiments, we determined that these compounds activated the complement system. Finally, we found that uptake of aggregation-prone iron nanomedicines by peripheral blood mononuclear cells in whole blood induced production of the proinflammatory cytokine IL-1ß, but not IL-6.


Subject(s)
Complement Activation/drug effects , Cytokines/immunology , Iron/administration & dosage , Leukocytes, Mononuclear/immunology , Metal Nanoparticles/administration & dosage , Toll-Like Receptors/immunology , Cells, Cultured , Complement Activation/immunology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Inflammation Mediators/immunology , Leukocytes, Mononuclear/drug effects , Up-Regulation/drug effects , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...