Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 188: 432-449, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34375663

ABSTRACT

Sulpiride (SUL), a benzamide derivative, acts as a multitarget drug with extensive biological properties. However, being a P-glycoprotein efflux substrate with a limited oral bioavailability imposes a challenge to its clinical efficacy. The current research explores the impact of tailored hybrid lipid-polysaccharide nanocomposites in augmenting the biological performance of SUL. Chitosan-graft-tocopherol polyethylene glycol 1000 succinate (TPGS) copolymers were synthesized and integrated as a polysaccharide shell into a SUL-loaded lipid nanocore. The optimized nanohybrids revealed a nanocore-shell structure with 110.1 nm particle size, 23.7 mV zeta potential, 85.42% encapsulation efficiency, a pH-dependent-release profile, and an acceptable mucoadhesive tendency. Employing TPGS into the chitosan backbone alleviated the cellular internalization of nanohybrids into the Caco-2 intestinal cells and hence increased the intestinal permeation and the oral bioavailability of SUL by 3.3, and 8.7-folds, respectively. Reserpine-induced depression rat model confirmed the superior antidepressant activity of nanohybrids, compared with free SUL and a marketed product. The nanohybrids exhibited 1.87- and 1.47-folds enhancement in both serotonin and dopamine levels, respectively. Additionally, nanohybrids were shown to attenuate brain oxidative stress state and SUL irritant effect on different body tissues. Overall, the newly tailored nanohybrids pave the way for an advance in the field of oral drug delivery.


Subject(s)
Chitosan/chemistry , Depressive Disorder/drug therapy , Nanocomposites/chemistry , Sulpiride/pharmacology , Administration, Oral , Caco-2 Cells , Chitosan/pharmacology , Humans , Lipids/chemistry , Lipids/pharmacology , Sulpiride/chemistry , Vitamin E/chemistry , Vitamin E/pharmacology
2.
Int J Nanomedicine ; 16: 4781-4803, 2021.
Article in English | MEDLINE | ID: mdl-34290503

ABSTRACT

BACKGROUND: Tacrolimus (TAC) is a powerful immunosuppressive agent whose therapeutic applicability is confined owing to its systemic side effects. OBJECTIVE: Herein, we harnessed a natural polymer based bioconjugate composed of maltodextrin and α-tocopherol (MD-α-TOC) to encapsulate TAC as an attempt to overcome its biological limitations while enhancing its therapeutic anti-rheumatic efficacy. METHODS: The designed TAC loaded maltodextrin-α-tocopherol nano-micelles (TAC@MD-α-TOC) were assessed for their physical properties, safety, toxicological behavior, their ability to combat arthritis and assist bone/cartilage formation. RESULTS: In vitro cell viability assay revealed enhanced safety profile of optimized TAC@MD-α-TOC with 1.6- to 2-fold increase in Vero cells viability compared with free TAC. Subacute toxicity study demonstrated a diminished nephro- and hepato-toxicity accompanied with optimized TAC@MD-α-TOC. TAC@MD-α-TOC also showed significantly enhanced anti-arthritic activity compared with free TAC, as reflected by improved clinical scores and decreased IL-6 and TNF-α levels in serum and synovial fluids. Unique bone formation criteria were proved with TAC@MD-α-TOC by elevated serum and synovial fluid levels of osteocalcin and osteopontin mRNA and proteins expression. Chondrogenic differentiation abilities of TAC@MD-α-TOC were proved by increased serum and synovial fluid levels of SOX9 mRNA and protein expression. CONCLUSION: Overall, our designed bioconjugate micelles offered an excellent approach for improved TAC safety profile with enhanced anti-arthritic activity and unique bone formation characteristics.


Subject(s)
Arthritis, Rheumatoid , Micelles , Animals , Bone Regeneration , Chlorocebus aethiops , Humans , Nanostructures , Polysaccharides , Tacrolimus , Tocopherols , Vero Cells
3.
Int J Nanomedicine ; 16: 2013-2044, 2021.
Article in English | MEDLINE | ID: mdl-33727812

ABSTRACT

BACKGROUND: Sulpiride (SUL), is a selective antidopaminergic drug that had extensive biological activities. However, its sparingly aqueous solubility and limited gastrointestinal permeability lead to scanty oral bioavailability which hinders its clinical efficacy. OBJECTIVE: SUL-loaded lipospheres (SUL-LPS) were designed to serve as an oral biocompatible nanovector for improving SUL permeability as well as conquering its low oral absorption and then in turn enhancing its antidepressant action. METHODS: SUL-LPS were fabricated via two processing techniques namely, melt emulsification and solvent evaporation. The impact of different lipid cores, phospholipid shells together with various surfactant concentrations and types on the lipospheres properties were screened. Detailed physicochemical elucidations were performed followed by ex vivo permeation appraisal using the non-everted intestine model. The pharmacokinetic parameters of SUL-LPS, free SUL and marketed product were assessed following oral administration to healthy rats. Reserpine-induced depression rat model was used to assess the antidepressant action of SUL-LPS on which full behavioural and biochemical analysis was conducted. Safety attributes of nanoencapsulated SUL on the brain and other internal organs were evaluated. RESULTS: The optimum LPS revealed an excellent nanosize with a narrow PdI, negative zeta potential and acceptable entrapment efficiency of 68.62 nm, 0.242, -30.4 mV and 84.12%, respectively. SUL-LPS showed a sustained release pattern and 2.1-fold enhancement in the intestinal permeation parameters with low mucin interaction. Oral pharmacokinetic appraisal exhibited that LPS provided 3.4-fold improvement in SUL oral bioavailability together with long-circulating properties, relative to the free drug. Pharmacodynamic study confirmed the superior antidepressant action of SUL-LPS as evident by 1.6 and 1.25-fold elevation in the serotonin and dopamine expressions, respectively. Meanwhile, nanotoxicological appraisal proved the biocompatibility of SUL-LPS upon repetitive oral administration. CONCLUSION: Rationally designed lipospheres hold promising in vitro and in vivo characteristics for efficient delivery of SUL with high oral bioavailability, antidepressant activity together with a good safety profile.


Subject(s)
Antidepressive Agents/pharmacology , Lipids/chemistry , Nanoparticles/chemistry , Sulpiride/administration & dosage , Sulpiride/pharmacology , Administration, Oral , Animals , Biocompatible Materials/chemistry , Biological Availability , Chromatography, High Pressure Liquid , Drug Compounding , Drug Liberation , Freeze Drying , Male , Mucins/chemistry , Nanoparticles/ultrastructure , Neurotransmitter Agents/metabolism , Organ Specificity/drug effects , Particle Size , Permeability , Rats, Sprague-Dawley , Rats, Wistar , Sulpiride/chemistry , Sulpiride/pharmacokinetics , Swine
4.
Int J Pharm ; 590: 119919, 2020 Nov 30.
Article in English | MEDLINE | ID: mdl-32991960

ABSTRACT

Diabetic ulcers are prone to bacterial contamination and can severely affect patient's quality of life. This study is first report to explore copper-grafted graphene oxide-crosslinked zein scaffolds (Cu-GZS) for promoting cutaneous excision wounds healing as a promising therapeutic modality in diabetic male-rats. Cu-GZS scaffolds were fabricated using electrospinning technique, where GO was employed as an echo-friendly crosslinker to meliorate mechanical stability and swellability of scaffolds. To circumvent risk of infection, copper ions were grafted into GZS as bactericidal agents and angiogenesis promoters, through soaking GZS nanofibers into Cu-solution or direct loading during electrospinning process. SEM images showed GO encapsulation by wrapping around/or trapping within nanofibrous. Interestingly, formation of chemical amide bond between zein and GO was proven by FTIR spectra. Crosslinking of zein nanofibers with GO increased tensile strength of nanofibrous by 3-folds compared to uncrosslinked zein nanofibers. Optimized Cu-GZS exhibited constant release rate of copper over a period of 8 days (~53.42%). Cu-GZS immensely accelerated wound closure demonstrated by diminished infiltration of leukocytes, absence of α-SMA positive cells, presence of fully intact epithelium with normal keratinization and accelerated wound size reduction, compared to control. Cu-GZS scaffolds could serve as promising biomaterials for effective topical wound healing in diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental , Nanofibers , Zein , Adsorption , Animals , Graphite , Male , Quality of Life , Rats , Tissue Scaffolds , Wound Healing
5.
Int J Nanomedicine ; 14: 8251-8270, 2019.
Article in English | MEDLINE | ID: mdl-31802865

ABSTRACT

OBJECTIVES: Novel α-bisabolol (BIS)-loaded citric acid cross-linked zein nanofibrous scaffolds (C-ZNFs) were proposed to serve as safe platforms for promoting wound repair in rats. METHODS: ZNFs were synthesized using electrospinning technique, then NFs, with adequate water resistance, were produced using citric acid as a safe cross-linker. RESULTS: Compared to the uncross-linked ZNFs, cross-linking with 7% w/w citric acid decreased swelling index by 3 folds, while the tensile strength and the contact angle were enhanced to 2.5 and 3.8 folds, respectively. SEM images showed beads-free homogeneous NFs with a fully inter-connected 3D-network, where the average diameter of optimized C-ZNFs was 181.7±50 nm. After 24 h, C-ZNFs exhibited a decreased BIS release rate (45.6%), compared to uncross-linked mats (84.9%). By increasing BIS concentration, the cell adhesion (WI38 fibroblasts) was improved which can be attributed mainly to BIS activation of transforming growth factor-beta (TGF-ß1). The MTT-OD obtained values indicated that all tested zein scaffolds significantly enhanced the viability of WI38 fibroblasts, compared to the control after 48h of incubation which can be referred to the proliferative potential of zein by provoking cell spreading process. The scratch wound assay demonstrated that BIS-loaded ZNF scaffolds showed accelerated migration and proliferation of fibroblasts expressed by significantly higher wound closure rates compared to the control sample. BIS-loaded-C-ZNFs prominently accelerated tissue regeneration for wound closure demonstrated by entirely grown epithelium with normal keratinization and rapid wound contraction, compared to the control. Immunohistochemical results confirmed the superiority of BIS-loaded-C-ZNFs, where the observed reduced NF-κB and the elevated cytokeratin expressions confirmed the anti-inflammatory and proliferative effects of the scaffolds, respectively. CONCLUSION: In-vitro, optimized C-ZNFs offered a satisfactory cytocompatibility, adhesion and healing which were consistent with the in-vivo results. BIS-loaded-C-ZNFs could be regarded as a promising and effective biomaterial for tissue regeneration and for accelerating the wound healing process.


Subject(s)
Cross-Linking Reagents/chemistry , Monocyclic Sesquiterpenes/pharmacology , Nanofibers/chemistry , Regeneration/drug effects , Tissue Scaffolds/chemistry , Wound Healing/drug effects , Zein/chemistry , Animals , Cell Line , Cell Survival/drug effects , Drug Liberation , Fibroblasts/drug effects , Fibroblasts/pathology , Humidity , Male , Nanofibers/ultrastructure , Rats, Sprague-Dawley , Spectroscopy, Fourier Transform Infrared , Temperature , Thermogravimetry , Wettability
6.
Curr Drug Deliv ; 13(6): 899-908, 2016.
Article in English | MEDLINE | ID: mdl-26563940

ABSTRACT

The use of different drug classes as host modulating agents has been postulated to have significance as an adjunctive remedy curing chronic periodontitis. In this study nano-structured films containing doxycyclin (DOX) were evaluated for such purpose. Nano DOX/chitosan particulate system was prepared using spray drying technique and was then incorporated in PVA-based films. The particles were evaluated for particle size, zeta potential and possible drug/polymer interaction. The films were also tested for in-vitro drug release and clinical efficacy compared with placebo and DOX-loaded films. The formed particles had a zeta potential of + 13.8 mV and particle size of 52.86 nm with a polydispersity of PDI=0.946. No significant drug/polymer interaction was detected by DSC thermal analysis. In-vitro DOX release was sustained for about a week with the nano-structured films showing 23% of the drug released compared with 44% released from DOX films. Clinical efficacy was done on 150 periodontal pockets from patients suffering from moderate chronic periodontitis. Following scaling and root planning they were divided into three groups; group I receiving nano-structured (DOX), group II receiving DOX and group III receiving placebo films. Evaluation was done both clinically and biochemically at base-line, 1 week, 1 month and 2 months following drug application. Clinical findings indicated a significant effect of both nano-structured and DOX films in improving the measured parameters compared with the control and placebo groups.


Subject(s)
Doxycycline/administration & dosage , Doxycycline/therapeutic use , Nanostructures/administration & dosage , Periodontitis/drug therapy , Chitosan/chemistry , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/therapeutic use , Doxycycline/chemistry , Drug Liberation , Female , Humans , Male , Nanostructures/therapeutic use , Particle Size , Periodontal Pocket/drug therapy
7.
Int J Pharm ; 491(1-2): 113-22, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26095913

ABSTRACT

In this study, spray-dried alfuzosin hydrochloride (ALF)-loaded casein (CAS) nanoparticles were successfully used for the preparation of a swellable floating matrix via direct compression. The developed NIR calibration model was able to assess ALF and CAS levels in five different batches of drug-loaded nanoparticles. The calibration and prediction plots exhibited good linearity with correlation coefficients of more than 0.9. The standard error of calibration and cross-validation was less than 5% of the measured values, confirming the accuracy of the model. A linear relationship was obtained correlating the actual drug entrapped and the predicted values obtained from the NIR partial least squares regression model. The un-crosslinked tablet demonstrated a substantial weight gain (317% after 2h) and completely disintegrated after 3-4h whereas both 10 and 40% w/w genipin-crosslinked tablets showed lower weight gain (114 and 42% after 2h, respectively). A rapid floating of the tablets within 5-15min (compared to 45min for the marketed tablet) was observed, with maintained floating for 24h. Marketed and prepared tablets succeeded to prolong ALF release for 24h. The development of drug-loaded CAS nanoparticles using spray-drying represents a new alternative for the preparation of swellable floating tablets for prolonged drug release.


Subject(s)
Caseins/chemistry , Nanoparticles/chemistry , Tablets/chemistry , Calibration , Chemistry, Pharmaceutical/methods , Drug Compounding/methods , Iridoids/chemistry , Spectroscopy, Near-Infrared/methods
8.
Expert Opin Drug Deliv ; 11(10): 1525-36, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24940608

ABSTRACT

PURPOSE: Study the possible benefit of combining biodegradable polymers with sildenafil citrate (SC) in wound healing. METHOD: Biodegradable micronized powdered formulations of SC were prepared by spray drying using chitosan (P1) or chitosan/gum Arabic (P2). Powders were characterized by differential scanning calorimetry, Scanning electron microscope, particle size analysis, flow and swelling behavior. The powders were also incorporated into microstructured gels and in vitro SC release from powders and gels was tested. In vivo wound healing acceleration was tested by measuring area contraction of excision wounds and histologically. Post-healing tensile strength (TS) for incision wounds in rats receiving powder formulations was tested. RESULTS: The powders were in the micron-size range showing no SC-polymers interaction. Powders had poor flow with angle of repose (θ) of 41 - 48°, and high moisture uptake reaching 107% for placebo powder P1. Good excision wound healing was seen with P1 and G1 formulations showing 98.4 and 98.5% reduction in wound area, respectively, compared with 83% for the control. Incision wounds were improved with P1 showing TS value of 6.9 compared with 3.7 kg/cm² for control. Histological examinations supported. CONCLUSION: Spray-dried chitosan/SC powder (P1) and its gel form (G1) could be promising wound healing promoters as supported by the histological examinations.


Subject(s)
Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Skin/drug effects , Sulfones/pharmacology , Wound Healing/drug effects , Animals , Calorimetry, Differential Scanning , Chemistry, Pharmaceutical , Chitosan/chemistry , Female , Microscopy, Electron, Scanning , Microspheres , Particle Size , Phosphodiesterase 5 Inhibitors/chemistry , Piperazines/chemistry , Powders , Purines/chemistry , Purines/pharmacology , Rats , Rats, Sprague-Dawley , Sildenafil Citrate , Sulfones/chemistry
9.
Eur J Pharm Biopharm ; 85(3 Pt A): 444-51, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23872177

ABSTRACT

In this study, casein (CAS) nanoparticles were used to encapsulate the hydrophobic anticancer drug, flutamide (FLT), aiming at controlling its release, enhancing its anti-tumor activity, and reducing its hepatotoxicity. The nanoparticles were prepared by emulsification of CAS, at pH below its isoelectric point, and stabilized via ionic-crosslinking with sodium tripolyphosphate (TPP). The nanoparticles were spherical and positively charged with a size below 100 nm and exhibited a sustained drug release up to 4 days. After intravenous administration into prostate cancer-bearing rats for 28 days, FLT-loaded CAS nanoparticles showed a higher anti-tumor efficacy as revealed by a significantly higher % reduction in PSA serum level (75%) compared to free FLT (55%). Moreover, the nanoparticles demonstrated a marked reduction in the relative weights of both prostate tumor and seminal vesicle (43% and 32%) compared to free FLT (12% and 18%), respectively. A significantly higher anti-proliferative, anti-angiogenic, and apoptotic effects was demonstrated by the nanoparticles compared to drug solution as evidenced by their ability to decrease the expression of the proliferative marker (Ki-67) and reduce the level of tumor angiogenic markers (VEGF and IGF-1) as well as their ability to activate caspase-3 with subsequent induction of apoptosis in prostate cancer cells. Conclusively, these novel ionically-crosslinked milk protein nanovehicles offer a promising carrier to allow controlled intravenous delivery of hydrophobic anticancer drugs.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Flutamide/administration & dosage , Nanoparticles , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents, Hormonal/chemistry , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis/drug effects , Caseins/chemistry , Caspase 3/metabolism , Cross-Linking Reagents/chemistry , Delayed-Action Preparations , Drug Carriers/chemistry , Drug Delivery Systems , Flutamide/pharmacology , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Male , Particle Size , Polyphosphates/chemistry , Prostatic Neoplasms/pathology , Rats , Rats, Sprague-Dawley
10.
Pharm Res ; 30(10): 2654-63, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23739989

ABSTRACT

PURPOSE: This article describes the preparation, physicochemical characterization and in vivo assessment of parenteral colloidal formulation of flutamide (FLT) based on biocompatible casein (CAS) self-assembled micelles in order to control drug release, enhance its antitumor efficacy and reduce its hepatotoxicity. METHODS: Spray-drying technique was successfully utilized to obtain solidified redispersible drug-loaded micelles. RESULTS: Spherical core-shell micelles were obtained with a particle size below 100 nm and a negative zeta potential above -30 mV exhibiting a sustained drug release up to 5 days. After intravenous administration into prostate cancer bearing rats for 28 days, FLT-loaded CAS micelles showed a higher antitumor efficacy as revealed by significantly higher reduction in PSA serum level (65.95%) compared to free FLT (55.43%). Moreover, micellar FLT demonstrated a marked decrease in relative weights of both prostate tumor and seminal vesicle (34.62 and 24.59%) compared to free FLT (11.86 and 17.74%), respectively. These antitumor responses were associated with notable reduction of cell proliferation, intratumoral angiogenesis and marked increase of tumor apoptosis. A significantly lower risk of hepatotoxicity was observed by micellar FLT as evidenced by lower alanine aminotransferase (ALT) serum level compared to free FLT. CONCLUSIONS: Overall this approach suggested that CAS micelles might be an ideal candidate for intravenous delivery of hydrophobic anticancer drugs.


Subject(s)
Androgens/metabolism , Antineoplastic Agents, Hormonal/administration & dosage , Caseins/chemistry , Drug Carriers/chemistry , Flutamide/administration & dosage , Nanoparticles/chemistry , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Delayed-Action Preparations , Drug Compounding , Flutamide/therapeutic use , Male , Micelles , Particle Size , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Rats , Rats, Sprague-Dawley , Solubility , Surface Properties
11.
Int J Nanomedicine ; 8: 1721-32, 2013.
Article in English | MEDLINE | ID: mdl-23658490

ABSTRACT

A novel particulate delivery matrix based on ionically crosslinked casein (CAS) nanoparticles was developed for controlled release of the poorly soluble anticancer drug flutamide (FLT). Nanoparticles were fabricated via oil-in-water emulsification then stabilized by ionic crosslinking of the positively charged CAS molecules below their isoelectric point, with the polyanionic crosslinker sodium tripolyphosphate. With the optimal preparation conditions, the drug loading and incorporation efficiency achieved were 8.73% and 64.55%, respectively. The nanoparticles exhibited a spherical shape with a size below 100 nm and a positive zeta potential (+7.54 to +17.3 mV). FLT was molecularly dispersed inside the nanoparticle protein matrix, as revealed by thermal analysis. The biodegradability of CAS nanoparticles in trypsin solution could be easily modulated by varying the sodium tripolyphosphate crosslinking density. A sustained release of FLT from CAS nanoparticles for up to 4 days was observed, depending on the crosslinking density. After intravenous administration of FLT-CAS nanoparticles into rats, CAS nanoparticles exhibited a longer circulation time and a markedly delayed blood clearance of FLT, with the half-life of FLT extended from 0.88 hours to 14.64 hours, compared with drug cosolvent. The results offer a promising method for tailoring biodegradable, drug-loaded CAS nanoparticles as controlled, long-circulating drug delivery systems of hydrophobic anticancer drugs in aqueous vehicles.


Subject(s)
Caseins/chemistry , Drug Carriers/pharmacokinetics , Flutamide/pharmacokinetics , Nanoparticles/chemistry , Animals , Delayed-Action Preparations , Drug Carriers/chemistry , Drug Carriers/pharmacology , Flutamide/chemistry , Flutamide/pharmacology , Male , Polyphosphates , Rats , Rats, Sprague-Dawley
12.
Eur J Pharm Biopharm ; 84(3): 487-96, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23403015

ABSTRACT

Novel casein (CAS)-based micelles loaded with the poorly soluble anti-cancer drug, flutamide (FLT), were successfully developed in a powdered form via spray-drying technique. Genipin (GNP) was used to crosslink CAS micelles as demonstrated by color variation of the micelles. Drug solubilization was enhanced by incorporation within the hydrophobic micellar core which was confirmed by solubility study and UV spectra. Spherical core-shell micelles were obtained with a particle size below 100 nm and zeta potential around -30 mV. At low drug loading, FLT was totally incorporated within micellar core as revealed by thermal analysis. However, at higher loading, excess non-incorporated drug at micelle surface caused a significant reduction in the surface charge density. Turbidity measurements demonstrated the high physical stability of micelles for 2 weeks dependent on GNP-crosslinking degree. In a dry powdered form, the micelles were stable for 6 months with no significant changes in drug content or particle size. A sustained drug release from CAS micelles up to 5 days was observed. After i.v. administration into rats, CAS micelles exhibited a prolonged plasma circulation of FLT compared to drug solution. Furthermore, a more prolonged drug systemic circulation was observed for GNP-crosslinked micelles. Overall, this study reports the application of spray-dried natural protein-based micelles for i.v. delivery of hydrophobic anti-cancer drugs such as FLT.


Subject(s)
Caseins/chemistry , Chemistry, Pharmaceutical/methods , Drug Delivery Systems , Flutamide/chemistry , Micelles , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Calorimetry, Differential Scanning , Chromatography, High Pressure Liquid , Cross-Linking Reagents/chemistry , Flutamide/pharmacokinetics , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Iridoids/chemistry , Male , Particle Size , Rats , Rats, Sprague-Dawley , Spectroscopy, Fourier Transform Infrared , Time Factors , Ultraviolet Rays
13.
Pharm Res ; 30(2): 512-22, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23135815

ABSTRACT

PURPOSE: To propose a simple method for the development of genipin-crosslinked casein micelles as a new delivery platform for prolonged release of alfuzosin hydrochloride. METHODS: Crosslinked casein micelles entrapping alfuzosin were transformed into solid redispersible nanoparticles via spray-drying technique with no need for drying adjuvants based on the stabilizing effect of casein. RESULTS: The nanoparticles displayed high production yields (86.99-94.63% w/w) with a reasonable drug incorporation efficiency ranged from 92.86 to 97.75%. The nanoparticles were readily reconstituted in aqueous solution with a particle size range of 122.1-260.0 nm and a zeta potential range of -21.6 to -36.6 mV indicating a good colloidal stability. No drug crystals were detectable in the scanning electron micrographs revealing successful encapsulation of alfuzosin into casein nanoparticles which was confirmed by differential scanning calorimetry. The nanoparticles succeeded in prolonging the drug release that could be controlled by modulating the genipin crosslinking degree. The release data showed a good fit into Higuchi release kinetics with non-Fickian type of drug diffusion. CONCLUSIONS: These results demonstrated that genipin-crosslinking combined with spray-drying technique could be used as a promising tool to develop solid redispersible casein nanoparticles with sustained drug release properties.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/administration & dosage , Caseins/chemistry , Delayed-Action Preparations/chemistry , Iridoids/chemistry , Nanoparticles/chemistry , Quinazolines/administration & dosage , Cross-Linking Reagents/chemistry , Desiccation/methods
14.
J Control Release ; 161(1): 38-49, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22564368

ABSTRACT

Among the available potential colloidal drug carrier systems, protein-based nanocarriers are particularly interesting. Meeting requirements such as low cytotoxicity, abundant renewable sources, high drug binding capacity and significant uptake into the targeted cells, protein-based nanocarriers represent promising candidates for efficient drug and gene delivery. Moreover, the unique protein structure offers the possibility of site-specific drug conjugation and targeting using various ligands modifying the surface of protein nanocarriers. The current review highlights the main advances achieved in utilizing protein nanocarriers as natural vehicles for drug and gene delivery tasks with respect to types, advantages, limitations, formulation aspects as well as the major outcomes of the in vitro and in vivo investigations. The recently emerged technologies in the formulation of protein nanocarriers including using recombinant proteins as alternatives to native ones and new non-toxic crosslinkers as alternatives to the toxic chemical crosslinkers are also discussed.


Subject(s)
Drug Carriers/chemistry , Gene Transfer Techniques , Nanostructures/chemistry , Proteins/chemistry , Animals , Drug Carriers/metabolism , Humans , Nanostructures/ultrastructure , Plant Proteins/chemistry , Plant Proteins/metabolism , Plants/chemistry , Plants/metabolism , Proteins/metabolism
15.
J Control Release ; 157(2): 168-82, 2012 Jan 30.
Article in English | MEDLINE | ID: mdl-21839127

ABSTRACT

Albumin, a versatile protein carrier for drug delivery, has been shown to be nontoxic, non-immunogenic, biocompatible and biodegradable. Therefore, it is ideal material to fabricate nanoparticles for drug delivery. Albumin nanoparticles have gained considerable attention owing to their high binding capacity of various drugs and being well tolerated without any serious side-effects. The current review embodies an in-depth discussion of albumin nanoparticles with respect to types, formulation aspects, major outcomes of in vitro and in vivo investigations as well as site-specific drug targeting using various ligands modifying the surface of albumin nanoparticles with special insights to the field of oncology. Specialized nanotechnological techniques like desolvation, emulsification, thermal gelation and recently nano-spray drying, nab-technology and self-assembly that have been investigated for fabrication of albumin nanoparticles, are also discussed. Nanocomplexes of albumin with other components in the area of drug delivery are also included in this review.


Subject(s)
Albumins/pharmacology , Drug Carriers/pharmacology , Nanoparticles , Albumins/chemistry , Animals , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Drug Carriers/chemistry , Humans , Nanoparticles/chemistry , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...