Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Int J Pharm ; 658: 124208, 2024 Jun 10.
Article En | MEDLINE | ID: mdl-38723731

Pseudomonas aeruginosa (PA), a predominant pathogen in lung infections, poses significant challenges due to its biofilm formation, which is the primary cause of chronic and recalcitrant pulmonary infections. Bacteria within these biofilms exhibit heightened resistance to antibiotics compared to their planktonic counterparts, and their secreted toxins exacerbate lung infections. Diverging from traditional antibacterial therapy for biofilm eradication, this study introduces a novel dry powder inhalation containing muco-inert ciprofloxacin and colistin co-encapsulated liposomes (Cipro-Col-Lips) prepared using ultrasonic spray freeze drying (USFD) technique. This USFD dry powder is designed to efficiently deliver muco-inert Cipro-Col-Lips to the lungs. Once deposited, the liposomes rapidly diffuse into the airway mucus, reaching the biofilm sites. The muco-inert Cipro-Col-Lips neutralize the biofilm-secreted toxins and simultaneously trigger the release of their therapeutic payload, exerting a synergistic antibiofilm effect. Our results demonstrated that the optimal USFD liposomal dry powder formulation exhibited satisfactory in vitro aerosol performance in terms of fine particle fraction (FPF) of 44.44 ± 0.78 %, mass median aerodynamic diameter (MMAD) of 4.27 ± 0.21 µm, and emitted dose (ED) of 99.31 ± 3.31 %. The muco-inert Cipro-Col-Lips effectively penetrate the airway mucus and accumulate at the biofilm site, neutralizing toxins and safeguarding lung cells. The triggered release of ciprofloxacin and colistin works synergistically to reduce the biofilm's antibiotic resistance, impede the development of antibiotic resistance, and eliminate 99.99 % of biofilm-embedded bacteria, including persister bacteria. Using a PA-beads induced biofilm-associated lung infection mouse model, the in vivo efficacy of this liposomal dry powder aerosol was tested, and the results demonstrated that this liposomal dry powder aerosol achieved a 99.7 % reduction in bacterial colonization, and significantly mitigated inflammation and pulmonary fibrosis. The USFD dry powder inhalation containing muco-inert Cipro-Col-Lips emerges as a promising therapeutic strategy for treating PA biofilm-associated lung infections.


Anti-Bacterial Agents , Biofilms , Ciprofloxacin , Colistin , Dry Powder Inhalers , Liposomes , Pseudomonas Infections , Pseudomonas aeruginosa , Ciprofloxacin/administration & dosage , Ciprofloxacin/pharmacology , Ciprofloxacin/chemistry , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Biofilms/drug effects , Colistin/administration & dosage , Colistin/pharmacology , Administration, Inhalation , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Pseudomonas Infections/drug therapy , Mice , Aerosols , Lung/microbiology , Lung/drug effects , Powders , Female , Particle Size
2.
Pharmaceutics ; 15(12)2023 Nov 30.
Article En | MEDLINE | ID: mdl-38140054

Gefitinib (GEF) is a clinical medication for the treatment of lung cancer targeting the epidermal growth factor receptor (EGFR). However, its efficacy is remarkably limited by low solubility and dissolution rates. In this study, two cocrystals of GEF with co-formers were successfully synthesized using the recrystallization method characterized via Powder X-ray Diffraction, Fourier Transform Infrared Spectroscopy, and 2D Nuclear Overhauser Effect Spectroscopy. The solubility and dissolution rates of cocrystals were found to be two times higher than those of free GEF. In vitro cytotoxicity studies revealed that the cocrystals enhanced the inhibition of cell proliferation and apoptosis in A549 and H1299 cells compared to free GEF. In mouse models, GEF@TSBO demonstrated targeted, safe, and effective antitumor activity with only one-dose administration. Mechanistically, the GEF cocrystals were shown to increase the cellular levels of damaged DNA, while potentially downregulating PARP, thereby impairing the DNA repair machinery and leading to an imbalance between DNA damage and restoration. These findings suggest that the cocrystallization of GEF could serve as a promising adjunct to significantly enhance the physicochemical and biopharmaceutical performance for lung cancer treatment, providing a facial strategy to improve GEF anticancer efficiency with high bioavailability that can be orally administrated with only one dose.

3.
Pharmaceutics ; 15(11)2023 Nov 03.
Article En | MEDLINE | ID: mdl-38004561

Airway mucus dysfunction and impaired immunological defenses are hallmarks of several lung diseases, including asthma, cystic fibrosis, and chronic obstructive pulmonary diseases, and are mostly causative factors in bacterial-biofilm-associated respiratory tract infections. Bacteria residing within the biofilm architecture pose a complex challenge in clinical settings due to their increased tolerance to currently available antibiotics and host immune responses, resulting in chronic infections with high recalcitrance and high rates of morbidity and mortality. To address these unmet clinical needs, potential anti-biofilm therapeutic strategies are being developed to effectively control bacterial biofilm. This review focuses on recent advances in the development and application of nanoparticulate drug delivery systems for the treatment of biofilm-associated respiratory tract infections, especially addressing the respiratory barriers of concern for biofilm accessibility and the various types of nanoparticles used to combat biofilms. Understanding the obstacles facing pulmonary drug delivery to bacterial biofilms and nanoparticle-based approaches to combatting biofilm may encourage researchers to explore promising treatment modalities for bacterial-biofilm-associated chronic lung infections.

4.
Pharmaceutics ; 15(5)2023 May 15.
Article En | MEDLINE | ID: mdl-37242742

Rheumatoid arthritis (RA) is an autoimmune disease of synovial inflammation that affects populations worldwide. Transdermal drug delivery systems for treating RA have increased but remain challenging. We fabricated a dissolving microneedle (MN) system with photothermal (PT) polydopamine (PDA) to co-load the non-steroidal anti-inflammatory drug loxoprofen (Lox) and the Janus kinase inhibitor tofacitinib (Tof), with the aim of co-delivering Lox and Tof directly to the articular cavity, aided by the combination of MN and PT. In vitro and in vivo permeation studies showed that the PT MN significantly promoted drug permeation and retention in the skin. An in vivo visualization of the drug distribution in the articular cavity showed that the PT MN significantly promoted drug retention in the articular cavity. Importantly, compared to the intra-articular injection of Lox and Tof, the application of the PT MN to a carrageenan/kaolin-induced arthritis rat model exhibited superior performance in reducing joint swelling, muscle atrophy, and cartilage destruction. Furthermore, the PT MN downregulated the mRNA expression levels of proinflammatory cytokines, including TNF-α, IL-1ß, iNOS, JAK2, JAK3, and STAT3. The results show that the PT MN transdermal co-delivery of Lox and Tof is a new synergetic therapy with high compliance and good therapeutic efficacy for RA.

5.
Theranostics ; 12(8): 3610-3627, 2022.
Article En | MEDLINE | ID: mdl-35664057

The development of activatable photosensitizers (aPSs) responding to tumor-specific biomarkers for precision photodynamic therapy (PDT) is urgently required. Due to the unique proteolytic activity and highly restricted distribution of tumor-specific enzymes, enzyme activatable photosensitizers display superior selectivity. Methods: Herein, a series of novel Fibroblast Activation Protein α (FAPα) activatable theranostic pro-photosensitizers were designed by conjugating the different N-terminal blocked FAPα-sensitive dipeptide substrates with a clinical PS, methylene blue (MB), through a self-immolative linker, which resulting in the annihilation of the photoactivity (fluorescence and phototoxicity). The best FAPα-responsive pro-photosensitizer was screened out through hydrolytic efficiency and blood stability. Subsequently, a series of in vitro and in vivo experiments were carried out to investigate the FAPα responsiveness and enhanced PDT efficacy. Results: The pro-photosensitizers could be effectively activated by tumor-specific FAPα in the tumor sites. After response to FAPα, the "uncaged" MB can recover its fluorescence and phototoxicity for tumor imaging and cytotoxic singlet oxygen (1O2) generation, eventually achieving accurate imaging-guided PDT. Simultaneously, the generated azaquinone methide (AQM) could serve as a glutathione (GSH) scavenger to rapidly and irreversibly weaken intracellular antioxidant capacity, realizing synergistic oxidative stress amplification and enhanced PDT effect. Conclusion: This novel FAPα activatable theranostic pro-photosensitizers allow for accurate tumor imaging and admirable PDT efficacy with minimal systemic side effects, offering great potential in clinical precision antitumor application.


Dermatitis, Phototoxic , Neoplasms , Photochemotherapy , Cell Line, Tumor , Dermatitis, Phototoxic/drug therapy , Endopeptidases , Glutathione/metabolism , Humans , Membrane Proteins , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Precision Medicine , Theranostic Nanomedicine/methods
6.
Article En | MEDLINE | ID: mdl-33713949

The complexity of Tobradex® ointment formulation (dexamethasone 0.1 wt% and tobramycin 0.3 wt%) and the high cost of pharmacokinetic (PK) studies in human aqueous humor may prevent generic drug companies from moving forward with a Tobradex®-equivalent product development. The in vitro drug release test would be an alternative approach for differentiating the generic formulations containing both dexamethasone (DEX) and tobramycin (TOB), and the results should be correlated with the in vivo ocular PK studies for further evaluation. To facilitate the in vivo ocular PK studies, a sensitive, rapid and specific liquid chromatography-tandem mass spectrometry (LC-MS/MS) method that can simultaneously quantify both DEX and TOB in rabbit ocular matrices including tear, aqueous humor and cornea was established and validated. The lower limit of quantification (LLOQ) was 1.5 ng/ml for DEX and 3 ng/ml for TOB with good precision and accuracy. Both intra- and inter-batch precisions were within ±15%, and the accuracy for all QCs was within the range of 85-115%. This new method was successfully applied for a pilot pharmacokinetic analysis of DEX and TOB in rabbit tears after topical administration of Tobradex® ointment.


Aqueous Humor/chemistry , Chromatography, Liquid/methods , Dexamethasone/analysis , Tandem Mass Spectrometry/methods , Tobramycin/analysis , Animals , Anti-Bacterial Agents/analysis , Anti-Bacterial Agents/pharmacokinetics , Cornea/chemistry , Dexamethasone/pharmacokinetics , Female , Linear Models , Male , Rabbits , Reproducibility of Results , Sensitivity and Specificity , Tears/chemistry , Tobramycin/pharmacokinetics
7.
J Pharm Anal ; 11(6): 732-738, 2021 Dec.
Article En | MEDLINE | ID: mdl-35028178

Inhaled antibiotics such as colistin and ciprofloxacin are increasingly used to treat bacterial lung infections in cystic fibrosis patients. In this study, we established and validated a new HPLC-MS/MS method that could simultaneously detect drug concentrations of ciprofloxacin, colistin and ivacaftor in rat plasma, human epithelial cell lysate, cell culture medium, and drug transport media. An aliquot of 200 µL drug-containing rat plasma or cell culture medium was treated with 600 µL of extraction solution (acetonitrile containing 0.1% formic acid and 0.2% trifluoroacetic acid (TFA)). The addition of 0.2% TFA helped to break the drug-protein bonds. Moreover, the addition of 0.1% formic acid to the transport medium and cell lysate samples could significantly improve the response and reproducibility. After vortexing and centrifuging, the sample components were analyzed by HPLC-MS/MS. The multiple reaction monitoring mode was used to detect the following transitions: 585.5-101.1 (colistin A), 578.5-101.1 (colistin B), 393.2-337.2 (ivacaftor), 332.2-314.2 (ciprofloxacin), 602.3-101.1 (polymyxin B1 as internal standard (IS)) and 595.4-101.1 (polymyxin B2 as IS). The running time of a single sample was only 6 min, making this a time-efficient method. Linear correlations were found for colistin A at 0.029-5.82 µg/mL, colistin B at 0.016-3.14 µg/mL, ivacaftor at 0.05-10.0 µg/mL, and ciprofloxacin at 0.043-8.58 µg/mL. Accuracy, precision, and stability of the method were within the acceptable range. This method would be highly useful for research on cytotoxicity, animal pharmacokinetics, and in vitro drug delivery.

8.
Am J Respir Cell Mol Biol ; 64(1): 69-78, 2021 01.
Article En | MEDLINE | ID: mdl-33095650

Mucus obstruction is a key feature of many inflammatory airway diseases. Neutrophil extracellular traps (NETs) are released upon neutrophil stimulation and consist of extracellular chromatin networks studded with cytotoxic proteins. When released in the airways, these NETs can become part of the airway mucus. We hypothesized that the extracellular DNA and/or oxidative stress (e.g., by the release of reactive oxygen species and myeloperoxidase during NETs formation in the airways) would increase mucus viscoelasticity. We collected human airway mucus from endotracheal tubes of healthy patients admitted for elective surgery and coincubated these samples with NETs from phorbol 12-myristate 13-acetate-stimulated neutrophils. Unstimulated neutrophils served as controls, and blocking experiments were performed with dornase alfa for extracellular DNA and the free radical scavenger dimethylthiourea for oxidation. Compared with controls, the coincubation of mucus with NETs resulted in 1) significantly increased mucus viscoelasticity (macrorheology) and 2) significantly decreased mesh pore size of the mucus and decreased movement of muco-inert nanoparticles through the mucus (microrheology), but 3) NETs did not cause visible changes in the microstructure of the mucus by scanning EM. Incubation with either dornase alfa or dimethylthiourea attenuated the observed changes in macrorheology and microrheology. This suggests that the release of NETs may contribute to airway mucus obstruction by increasing mucus viscoelasticity and that this effect is not solely due to the release of DNA but may in part be due to oxidative stress.


Extracellular Traps/immunology , Mucus/immunology , Neutrophils/immunology , Respiratory System/immunology , Adult , Airway Obstruction/immunology , Airway Obstruction/metabolism , Extracellular Traps/metabolism , Humans , Mucus/metabolism , Neutrophils/metabolism , Oxidative Stress/immunology , Peroxidase/immunology , Peroxidase/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Respiratory System/metabolism
9.
J Control Release ; 327: 456-466, 2020 11 10.
Article En | MEDLINE | ID: mdl-32822742

Corneal neovascularization (NV) predisposes patients to compromised corneal transparency and visional acuity. Sunitinib malate (Sunb-malate) targeting against multiple receptor tyrosine kinases, exerts potent antiangiogenesis. However, the rapid clearance of Sunb-malate eye drops administered through topical instillation limits its therapeutic efficacy and poses a challenge for potential patient compliance. Sunb-malate, the water-soluble form of sunitinib, was shown to have higher intraocular penetration through transscleral diffusion following subconjunctival (SCT) injection in comparison to its sunitinib free base formulation. However, it is difficult to load highly water-soluble drugs and achieve sustained drug release. We developed Sunb-malate loaded poly(D,L-lactic-co-glycolic acid) (PLGA) microspheres (Sunb-malate MS) with a particle size of approximately 15 µm and a drug loading of 7 wt%. Sunb-malate MS sustained the drug release for 30 days under the in vitro infinite sink condition. Subconjunctival (SCT) injection of Sunb-malate MS provided a prolonged ocular drug retention and did not cause ocular toxicity at a dose of 150 µg of active agent. Sunb-malate MS following SCT injection more effectively suppressed the suture-induced corneal NV than either Sunb-malate free drug or the placebo MS. Local sustained release of Sunb-malate through the SCT injection of Sunb-malate MS mitigated the proliferation of vascular endothelial cells and the recruitment of mural cells into the cornea. Moreover, the gene upregulation of proangiogenic factors induced by the pathological process was greatly neutralized by SCT injection of Sunb-malate MS. Our findings provide a sustained release platform for local delivery of tyrosine kinase inhibitors to treat corneal NV.


Corneal Neovascularization , Animals , Corneal Neovascularization/prevention & control , Drug Liberation , Endothelial Cells , Humans , Microspheres , Rats , Sunitinib
10.
Nanomedicine ; 29: 102262, 2020 10.
Article En | MEDLINE | ID: mdl-32623017

Tenacious sputum poses a critical diffusion barrier for aerosol antibiotics used to treat cystic fibrosis (CF) lung infection. We conducted a proof-of-concept study using dense poly(ethylene glycol) coated polystyrene nanoparticles (PS-PEG NPs) as model muco-inert particles (MIPs) formulated as a powder using an excipient enhanced growth (EEG) strategy, aiming to minimize extrathoracic airway loss, maximize deposition in the airway and further overcome the sputum barrier in the CF lungs. The EEG aerosol formulation containing PS-PEG MIPs was prepared by spray drying and produced discrete spherical particles with geometric diameter of approximately 2 µm; and >80% of the powder dose was delivered from a new small-animal dry powder inhaler (DPI). The MIPs released from the EEG aerosol had human airway mucus and CF sputum diffusion properties comparable to the suspension formulation. These properties make this formulation a promising pulmonary drug delivery system for CF lung infections.


Cystic Fibrosis/drug therapy , Drug Delivery Systems , Lung Diseases/drug therapy , Lung/drug effects , Nanoparticles/chemistry , Administration, Inhalation , Cystic Fibrosis/pathology , Dry Powder Inhalers/methods , Excipients/chemistry , Humans , Lung/growth & development , Lung Diseases/pathology , Mucus/drug effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polystyrenes/chemistry , Polystyrenes/pharmacology
11.
Int J Pharm ; 586: 119570, 2020 Aug 30.
Article En | MEDLINE | ID: mdl-32593649

Inhalation therapy has advantages for the treatment of multidrug resistant bacterial lung infections with high drug concentrations at the infection sites in the airways and reduced systemic exposure. We have developed liposomal formulations for pulmonary delivery of synergistic ciprofloxacin (Cipro) and colistin (Col) as the potential candidate for treatment of lung infections caused by multidrug resistant Gram-negative bacteria. This study aims to: (1) further optimize the powder formulation by adding drying stabilizers (polyvinyl pyrrolidone or poloxamer) to protect the liposomes during spray-freeze-drying; (2) evaluate the transport and cellular uptake of drugs in a human lung epithelial Calu-3 cell model. The liposomal powder formulations were produced using the ultrasonic spray-freeze-drying technique. The optimal formulation (F5) used mannitol (8% w/v) and sucrose (2% w/v) as the internal lyoprotectants. Adding external lyoprotectants/aerosolization enhancers (i.e. 8% w/v mannitol, 2% w/v sucrose and 1%, w/w PVP 10) produced the superior rehydrated EE values of ciprofloxacin and colistin (50.2 ± 0.9% for Cipro and 37.8 ± 1.2% for Col) as well as satisfactory aerosol performance (FPF: 34.2 ± 0.8% for Cipro and 33.6 ± 0.9% for Col). The cytotoxicity study indicated that F5 with the colistin concentration at 50 µg/mL and ciprofloxacin at 200 µg/mL was not cytotoxic to human lung epithelial Calu-3 cells. The intracellular uptake of ciprofloxacin was concentration-dependent in Calu-3 cells and the uptake of A-B was more than that of B-A for all samples (p < 0.05). This study demonstrates that co-delivery of ciprofloxacin and colistin in a single liposome can lower the transport capability of both drugs across the Calu-3 cell monolayer and their accumulation in the cells. These findings indicate that co-loaded liposomal powder of ciprofloxacin and colistin is a promising potential treatment for respiratory infections caused by multidrug resistant Gram-negative bacteria.


Anti-Bacterial Agents/administration & dosage , Ciprofloxacin/administration & dosage , Colistin/administration & dosage , Drug Delivery Systems , Administration, Inhalation , Aerosols , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/toxicity , Cell Line , Chemistry, Pharmaceutical , Ciprofloxacin/pharmacokinetics , Ciprofloxacin/toxicity , Colistin/pharmacokinetics , Colistin/toxicity , Drug Combinations , Epithelial Cells/metabolism , Humans , Liposomes , Lung/cytology , Lung/metabolism , Powders
12.
Int J Pharm ; 575: 118915, 2020 Feb 15.
Article En | MEDLINE | ID: mdl-31816354

The aim of this study was to design and characterize dry powder inhaler formulations of ciprofloxacin and colistin co-loaded liposomes prepared by the ultrasonic spray-freeze-drying (USFD) technique. Liposomal formulations and powder production parameters were optimized to achieve optimal characteristics and in-vitro performance such as encapsulation efficiency (EE), particle size, particle distribution index (PDI), fine particle fraction (FPF), emitted dose (ED) and in vitro antibacterial activity. The formulation (F6) with the mannitol (5% w/v) as the internal lyoprotectant and sucrose (5%, w/v), mannitol (10%, w/v) and leucine (5%, w/w) as the external lyoprotectants/aerosolization enhancers showed an optimal rehydrated EE values of ciprofloxacin and colistin (44.9 ± 0.9% and 47.0 ± 0.6%, respectively) as well as satisfactory aerosol performance (FPF: 45.8 ± 2.2% and 43.6 ± 1.6%, respectively; ED: 97.0 ± 0.5% and 95.0 ± 0.6%, respectively). For the blank liposomes, there was almost no inhibitory effect on the cell proliferation in human lung epithelial A549 cells, showing that the lipid materials used in the liposome formulation is safe for use in pulmonary drug delivery. The cytotoxicity study demonstrated that the optimized liposomal formulation (F6) was not cytotoxic at least at the drug concentrations of colistin 5 µg/mL and ciprofloxacin 20 µg/mL. Colistin (2 mg/L) monotherapy showed no antibacterial effect against P. aeruginosa H131300444 and H133880624. Ciprofloxacin (8 mg/L) monotherapy showed moderate bacterial killing for both clinical isolates; however, regrowth was observed in 6 h for P. aeruginosa H133880624. The liposomal formulation displayed superior antibacterial activity against clinical isolates of Pseudomonas aeruginosa H131300444 and P. aeruginosa H133880624 compared to each antibiotic per se. These results demonstrate that the liposomal powder formulation prepared by USFD could potentially be a pulmonary delivery system for antibiotic combination to treat multi-drug resistant Gram-negative lung infections.


Anti-Bacterial Agents/administration & dosage , Ciprofloxacin/administration & dosage , Colistin/administration & dosage , Gram-Negative Bacterial Infections/drug therapy , Respiratory Tract Infections/drug therapy , A549 Cells , Administration, Inhalation , Drug Combinations , Drug Resistance, Bacterial , Drug Resistance, Multiple , Drug Synergism , Humans , Liposomes , Powders , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development
13.
Int J Pharm ; 569: 118616, 2019 Oct 05.
Article En | MEDLINE | ID: mdl-31415873

Respiratory tract infections caused by multidrug-resistant Gram-negative bacteria are serious burdens to the public. Our previous findings indicated that co-loading of colistin and ciprofloxacin via liposomes improved in vitro antimicrobial activities against multidrug resistant Pseudomonas aeruginosa as compared to the monotherapies. The current study aims to investigate the transport behavior of colistin and ciprofloxacin in liposomes using the in vitro Calu-3 cell monolayer, which is a lung epithelial model cultured under the air-interfaced condition. The cell viability results demonstrated that there was no obvious toxicity of cells exposed to single or co-administered drugs at the concentration ≤500 µg/mL. Transport of ciprofloxacin into the cells was easier than that of colistin, which reached a plateau rapidly. Colistin was less trapped in the mucus or adhered to the apical cell membrane, and less transported across the cell monolayer than ciprofloxacin. The deposition of ciprofloxacin on the apical side increased over time (from 1 to 4 h). There was no drug-drug interaction observed during the transport of ciprofloxacin and colistin across the cell monolayer, when they were dosed together in the solution form. The amount of drug transported across the cell monolayer was decreased in both agents when loaded in liposomes. Both drugs were more trapped in the mucus or more adhered to the apical side cell membrane of the cell monolayer when they were in liposomes. This study demonstrated that co-delivery of colistin and ciprofloxacin in a single liposome can reduce transport capacity of both drugs across the lung epithelial cell monolayer and enhance drug retention on the lung epithelial surfaces; therefore, it is a promising approach to treat the respiratory infections caused by multidrug resistant Pseudomonas aeruginosa.


Anti-Bacterial Agents/administration & dosage , Ciprofloxacin/administration & dosage , Colistin/administration & dosage , Epithelial Cells/metabolism , Lung/cytology , Cell Line , Humans , Liposomes , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa
14.
Pharm Res ; 35(10): 187, 2018 Aug 09.
Article En | MEDLINE | ID: mdl-30094660

PURPOSE: This study aims to develop liposomal formulations containing synergistic antibiotics of colistin and ciprofloxacin for the treatment of infections caused by multidrug-resistant Pseudomonas aeruginosa. METHODS: Colistin (Col) and ciprofloxacin (Cip) were co-encapsulated in anionic liposomes by ammonium sulfate gradient. Particle size, encapsulation efficiency, in vitro drug release and in vitro antibiotic activities were evaluated. RESULTS: The optimized liposomal formulation has uniform sizes of approximately 100 nm, with encapsulation efficiency of 67.0% (for colistin) and 85.2% (for ciprofloxacin). Incorporation of anionic lipid (DMPG) markedly increased encapsulation efficiency of colistin (from 5.4 to 67.0%); however, the encapsulation efficiency of ciprofloxacin was independent of DMPG ratio. Incorporation of colistin significantly accelerated the release of ciprofloxacin from the DMPG anionic liposomes. In vitro release of ciprofloxacin and colistin in the bovine serum for 2 h were above 70 and 50%. The cytotoxicity study using A549 cells showed the liposomal formulation is as non-toxic as the drug solutions. Liposomal formulations of combinations had enhanced in vitro antimicrobial activities against multidrug resistant P. aeruginosa than the monotherapies. CONCLUSIONS: Liposomal formulations of two synergistic antibiotics was promising against multidrug resistant P. aeruginosa infections.


Anti-Infective Agents/pharmacology , Ciprofloxacin/pharmacology , Colistin/pharmacology , Phospholipids/chemistry , Pseudomonas aeruginosa/drug effects , A549 Cells , Anti-Infective Agents/chemistry , Cell Survival/drug effects , Ciprofloxacin/chemistry , Colistin/chemistry , Drug Compounding , Drug Resistance, Bacterial , Drug Synergism , Humans , Phosphatidylcholines/chemistry , Surface Properties
15.
ACS Appl Mater Interfaces ; 8(9): 5929-40, 2016 Mar 09.
Article En | MEDLINE | ID: mdl-26860241

Solid lipid nanoparticles (SLNs) have been extensively investigated and demonstrated to be a potential nanocarriers for improving oral bioavailability of many drugs. However, the molecular mechanisms related to this discovery are not yet understood. Here, the molecular transport mechanisms of the SLNs crossing simulative intestinal epithelial cell monolayers (Caco-2 cell monolayers) were studied. The cytotoxicology results of the SLNs in Caco-2 cells demonstrated that the nanoparticles had low cytotoxicity, had no effect on the integrity of the cell membrane, did not induce oxidative stress, and could significantly reduce cell membrane fluidity. The endocytosis of the SLNs was time-dependent, and their delivery was energy-dependent. For the first time, the transport of the SLNs was directly verified to be a vesicle-mediated process. The internalization of the SLNs was mediated by macropinocytosis pathway and clathrin- and caveolae (or lipid raft)-related routes. Transferrin-related endosomes, lysosomes, endoplasmic reticulum (ER), and Golgi apparatus were confirmed to be the main destinations of the SLNs in Caco-2 cells. As for the transport of the SLNs in Caco-2 cell monolayers, the results demonstrated that the SLNs transported to the basolateral side were intact, and the transport of the nanoparticles did not destroy the structure of tight junctions. The transcytosis of the SLNs across the Caco-2 cell monolayer was demonstrated to be mediated by the same routes as that in the endocytosis study. The ER, Golgi apparatus, and microtubules were confirmed to be important for the transport of the SLNs to both the basolateral and apical membrane sides. This study provides a more thoroughly understand of SLNs transportation crossing intestinal epithelial cell monolayers and could be beneficial for the fabrication of SLNs.


Cell Membrane/metabolism , Lipids/chemistry , Nanoparticles/chemistry , Caco-2 Cells , Cell Survival/drug effects , Endocytosis/drug effects , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Fluorescence Recovery After Photobleaching , Gold/chemistry , Golgi Apparatus/metabolism , Humans , Lysosomes/metabolism , Nanoparticles/metabolism , Nanoparticles/toxicity , Reactive Oxygen Species/metabolism
16.
Int J Nanomedicine ; 10: 5671-85, 2015.
Article En | MEDLINE | ID: mdl-26388691

PURPOSE: Nanoparticles (NPs) that target bone tissue were developed using poly(lactic-co-glycolic acid) (PLGA) copolymers and tetracycline (TC)-based bone-targeting moieties. These NPs are expected to enable the transport of drugs, such as simvastatin (SIM), for the treatment of osteoporosis. METHODS: The molecular structures of TC-PLGA were validated by (1)H-NMR, and the SIM-loaded NPs were prepared using the solvent emulsification method. The surface properties, cytotoxicity, cellular uptake, cell mineralization, bone targeting potential, and animal pharmacodynamics of the TC-PLGA NPs were evaluated and compared to those of PLGA NPs. RESULTS: It was confirmed that the average particle size of the NPs was approximately 220 nm. In phosphate-buffered saline (PBS, pH 7.4), the SIM-loaded NPs exhibited a cumulative release of up to 80% within 72 hours. An in vitro cell evaluation indicated that the NPs had an excellent cellular uptake capacity and showed great biocompatibility with MC3T3-E1 cells, thereby reducing the cytotoxic effects of SIM. The cell mineralization assay showed that the SIM-loaded NPs induced osteogenic differentiation and mineralized nodule formation in MC3T3-E1 cells, thereby achieving the same effect as SIM. Preliminary findings from in vitro and in vivo bone affinity assays indicated that the TC-PLGA NPs may display increased bone-targeting efficiency compared to PLGA NPs lacking a TC moiety. The use of SIM-loaded TC-PLGA NPs in treating osteoporosis was tested through animal pharmacodynamics analyses performed in ovariectomized rats, and the results suggested that the SIM-loaded TC-PLGA NPs can improve the curative effects of SIM on the recovery of bone mineral density compared to either SIM-loaded PLGA NPs or SIM alone. CONCLUSION: Bone-targeting NPs, which were based on the conjugation of TC to PLGA copolymers, have the ability to target bone. These NPs may be developed as a delivery system for hydrophobic drugs, and they are expected to improve the curative effects of drugs, reduce the administered drug doses, and reduce side effects in other organs.


Bone and Bones/drug effects , Drug Delivery Systems , Lactic Acid/chemistry , Nanoparticles/chemistry , Osteogenesis/drug effects , Polyglycolic Acid/chemistry , Simvastatin/chemistry , Tetracycline/chemistry , 3T3 Cells , Animals , Cell Differentiation , Chromatography, High Pressure Liquid , Female , Fluorescein/chemistry , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred ICR , Osteoporosis/drug therapy , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley , Solvents , Surface Properties
17.
Mol Pharm ; 11(10): 3716-26, 2014 Oct 06.
Article En | MEDLINE | ID: mdl-25197948

An understanding of drug delivery system transport across epithelial cell monolayer is very important for improving the absorption and bioavailability of the drug payload. The mechanisms of epithelial cell monolayer transport for various nanocarriers may differ significantly due to their variable components, surface properties, or diameter. Solid lipid nanoparticles (SLNs), conventionally formed by lipid materials, have gained increasing attention in recent years due to their excellent biocompatibility and high oral bioavailability. However, there have been few reports about the mechanisms of SLNs transport across epithelial cell monolayer. In this study, the molecular mechanisms utilized by SLNs of approximately 100 nm in diameter crossing intestinal epithelial monolayer were carefully studied using a simulative intestinal epithelial monolayer formed by Madin-Darby canine kidney (MDCK) epithelial cells. The results demonstrated that SLNs transportation did not induce a significant change on tight junction structure. We found that the endocytosis of SLNs into the epithelial cells was energy-dependent and was significantly greater than nanoparticle exocytosis. The endocytosis of SLNs was found to be rarely mediated via macropinocytosis, as confirmed by the addition of 5-(N-ethyl-N-isopropyl)amiloride (EIPA) as an inhibitory agent, and mainly depended on lipid raft/caveolae- and clathrin-mediated pathways. After SLNs was internalized into MDCK cells, lysosome was one of the main destinations for these nanoparticles. The exocytosis study indicated that the endoplasmic reticulum, Golgi complex, and microtubules played important roles in the transport of SLNs out of MDCK cells. The transcytosis study indicated that only approximately 2.5% of the total SLNs was transported from the apical side to the basolateral side. For SLNs transportation in MDCK cell monolayer, greater transport (approximately 4-fold) was observed to the apical side than to the basolateral side. Our findings may present a more comprehensive understanding on the transport of SLNs across epithelial cell monolayer.


Nanoparticles/chemistry , Nanoparticles/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Biological Transport , Cell Line , Dogs , Endocytosis/physiology , Exocytosis/physiology , Madin Darby Canine Kidney Cells
18.
Mol Pharm ; 10(5): 1865-73, 2013 May 06.
Article En | MEDLINE | ID: mdl-23495754

The aim of the present study was to evaluate the potential of PEGylated solid lipid nanoparticle (pSLN) as mucus penetrating particles (MPP) for oral delivery across gastrointestinal mucus. The SLN was prepared by an aqueous solvent diffusion method, subsequently modified with PEG2000-stearic acid (PEG2000-SA) as hydrophilic groups. Surface properties, cytotoxicity, cellular uptake, and transport across Caco-2/HT29 coculture cell monolayers, intestinal absorption, and pharmacokinetics of pSLN were studied compared with that of SLN. Quantitative cellular uptake showed that the internalization of SLN and pSLN was an active transfer process, which would be restrained by several inhibitors of cell activity. Compared with SLN, the permeation ability of pSLN decreased through Caco-2 cell monolayer while it increased through a mucus-secreting Caco-2/HT29 coculture cell monolayer, which indicated that the mucus layer has a significant impact on determining the efficiency of oral nanoformulations. In addition to increasing permeation ability, the stability of the nanoparticles in simulated intestinal fluids was also increased by the PEGylation. Moreover, in vitro everted gut sac technique and the ligated intestinal loops model in vivo also demonstrated that pSLN can rapidly penetrate mucus secretions, whereas the SLN were strongly trapped by highly viscoelastic mucus barriers. The pharmacokinetic studies presented that pSLN exhibited improved absorption efficiency and prolonged blood circulation times with a 1.99-fold higher relative bioavailability compared with SLN. In conclusion, PEGylated solid lipid nanoparticles had advantages in enhancing the bioavailability of oral administration.


Intestinal Absorption , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Coculture Techniques , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems , Endocytosis/drug effects , HT29 Cells , Humans , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Lipids/chemistry , Male , Nanoparticles/ultrastructure , Polyethylene Glycols/chemistry , Rats , Rats, Sprague-Dawley
19.
Drug Dev Ind Pharm ; 39(5): 646-56, 2013 May.
Article En | MEDLINE | ID: mdl-22583006

The main purpose of this study is to investigate the influence of two polysaccharides (dextran, hydroxyethyl starch) on the stability of parenteral emulsions. All parenteral emulsions were prepared by high-pressure homogenization. The influence of polysaccharides concentration was studied. The stabilities of autoclaving sterilization, centrifugation and freeze-thawing process were investigated extensively. Following the addition of polysaccharides, the stabilities of the parenteral emulsions were improved. A high-concentration polysaccharides solution (13%, w/v) produced better protection than a low one (1.3%, w/v), especially during freeze-thawing process. The protective mechanisms of polysaccharides were attributed to increasing systematic viscosity, non-frozen water absorbed by polysaccharides, formation of a linear bead-like structure and thicker mixed emulsifier film. Overall, polysaccharides can offer greatly increased protection for parenteral emulsions, and represent a novel protective strategy for improving the stability of this delivery system.


Dextrans/chemistry , Drug Stability , Emulsifying Agents/chemistry , Hydroxyethyl Starch Derivatives/chemistry , Infusions, Parenteral , Surface-Active Agents/chemistry , Centrifugation/methods , Chemistry, Pharmaceutical , Emulsions/chemistry , Freezing , Parenteral Nutrition , Pharmaceutical Preparations/chemistry , Plasma Substitutes/chemistry , Sterilization/methods
20.
Drug Dev Ind Pharm ; 36(9): 1054-63, 2010 Sep.
Article En | MEDLINE | ID: mdl-20818965

OBJECTIVE: The purpose of this study was to investigate the dissolution and oral bioavailability of an immediate-release tablet involving wet grinding of a poorly water-soluble drug, fenofibrate. METHODS: The milled suspension was prepared using a Basket Dispersing Mill in the presence of a hydrophilic polymer solution and then granulated with common excipients, and compressed into an immediate-release tablet with blank microcrystalline cellulose granules. RESULTS: Compared with unmilled tablets (56% within 30 minutes), the dissolution of wet-milled tablets (about 98% in 30 minutes) was markedly enhanced. No significant decrease in the dissolution rate (96% in 30 minutes) of the wet-milled tablet was observed after 3 months under 40 degrees C and 75% relative humidity storage. In addition, the oral bioavailability of the wet-milled tablets (test) and Lipanthyl supra-bioavailability tablets (reference) was determined in beagle dogs after a single dose (160 mg fenofibrate) in a randomized crossover, own-control study. The results suggested that both the area under the plasma concentration-time curve (AUC((0-t)) = 46.83 +/- 11.09 microg/mL h) and the mean peak concentration of the test (C(max) = 4.63 +/- 1.71 microg/mL) were higher than the reference (AUC((0-t)) = 35.12 +/- 10.97 microg/mL h, C(max) = 2.11 +/- 0.08 microg/mL). The relative bioavailability of the wet-milled tablet was approximately 1.3-fold higher. Furthermore, the apparent rate of absorption of fenofibrate from the wet-milled tablet (T(max) = 2.63 hours) was faster than that from Lipanthyl (T(max) = 3.75 hours). CONCLUSION: These results indicated that the dissolution and the bioavailability of fenofibrate were significantly enhanced by wet-grinding process. So, this shows that wet grinding is a powerful technique to improve the bioavailability for poorly water-soluble drugs, especially for Biopharmaceutics Classification System Class II compounds.


Fenofibrate/chemistry , Hypolipidemic Agents/chemistry , Administration, Oral , Animals , Biological Availability , Dogs , Drug Carriers/chemistry , Drug Compounding , Drug Stability , Excipients , Fenofibrate/pharmacokinetics , Hypolipidemic Agents/pharmacokinetics , Hypromellose Derivatives , Methylcellulose/analogs & derivatives , Methylcellulose/chemistry , Particle Size , Solubility , Suspensions , Tablets/chemistry , Therapeutic Equivalency
...