Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Biomed Mater Res B Appl Biomater ; 106(3): 1383-1413, 2018 04.
Article in English | MEDLINE | ID: mdl-28650116

ABSTRACT

Oral cancer is among the most prevalent cancers in the world. Moreover, it is one of the major health problems and causes of death in many regions of the world. The traditional treatment modalities include surgical removal, radiation therapy, systemic chemotherapy, or a combination of these methods. In recent decades, there has been significant interest in intraoral site-specific chemoprevention via local drug delivery using polymeric systems. Because of its easy accessibility and clear visibility, the oral mucosa is amenable for local drug delivery. A variety of polymeric systems-such as gels, tablets, films, patches, injectable systems (e.g., millicylindrical implants, microparticles, and in situ-forming depots), and nanosized carriers (e.g., polymeric nanoparticles, nanofibers, polymer-drug conjugates, polymeric micelles, nanoliposomes, nanoemulsions, and polymersomes)-have been developed and evaluated for the local delivery of natural and synthetic chemopreventive agents. The findings of in vitro, ex vivo, and in vivo studies and the positive outcome of clinical trials demonstrate that intraoral site-specific drug delivery is an attractive, highly effective and patient-friendly strategy for the management of oral cancer. Intraoral site-specific drug delivery provides unique therapeutic advantages when compared to systemic chemotherapy. Moreover, intraoral drug delivery systems are self-administrable and can be removed when needed, increasing patient compliance. This article covers important aspects and advances related to the design, development, and efficacy of polymeric systems for intraoral site-specific drug delivery. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1383-1413, 2018.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Drug Delivery Systems/methods , Mouth Neoplasms/prevention & control , Polymers/chemistry , Administration, Mucosal , Animals , Humans , Mouth Mucosa , Nanoparticles
2.
Mol Pharm ; 14(9): 3228-3237, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28726424

ABSTRACT

Herein we describe a formulation of self-encapsulating poly(lactic-co-glycolic acid) (PLGA) microspheres for vaccine delivery. Self-healing encapsulation is a novel encapsulation method developed by our group that enables the aqueous loading of large molecules into premade PLGA microspheres. Calcium phosphate (CaHPO4) adjuvant gel was incorporated into the microspheres as a protein-trapping agent for improved encapsulation of antigen. Microspheres were found to have a median size of 7.05 ± 0.31 µm, with a w/w loading of 0.60 ± 0.05% of ovalbumin (OVA) model antigen. The formulation demonstrated continuous release of OVA over a 49-day period. Released OVA maintained its antigenicity over the measured period of >21 days of release. C57BL/6 mice were immunized via the intranasal route with prime and booster doses of OVA (10 µg) loaded into microspheres or coadministered with cholera toxin B (CTB), the gold standard of mucosal adjuvants. Microspheres generated a Th2-type response in both serum and local mucosa, with IgG antibody responses approaching those generated by CTB. The results suggest that this formulation of self-encapsulating microspheres shows promise for further study as a vaccine delivery system.


Subject(s)
Lactic Acid/chemistry , Microspheres , Polyglycolic Acid/chemistry , Vaccines/administration & dosage , Vaccines/chemistry , Animals , Calcium Phosphates/chemistry , Cholera Toxin/chemistry , Chromatography, High Pressure Liquid , Drug Delivery Systems/methods , Enzyme-Linked Immunosorbent Assay , Female , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Ovalbumin/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer
3.
Am J Transl Res ; 8(6): 2693-704, 2016.
Article in English | MEDLINE | ID: mdl-27398152

ABSTRACT

Cell and growth factor-based tissue engineering has shown great potentials for skeletal regeneration. This study tested its feasibility in reconstructing large mandibular defects and compared the efficacy of varied construction materials and sealing methods. Bilateral mandibular critical-size (5-cm(3)) defects were created on six 4-month-old domestic pigs, and grafted with ß-tricalcium phosphate (ßTCP) only (Group-A), ßTCP with autologous bone marrow-derived mesenchymal stem cells (BM-MSCs) (Group-B), and ßTCP with BM-MSCs and biodegradable poly(lactic-co-glycolic acid) (PLGA) microspheres containing bone morphogenetic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF) (Group-C). The buccal sides of Groups-B/-C were either sealed by fibrin sealant or by a biodegradable PLGA barrier membrane before soft-tissue closure. Computed tomography (CT), microCT and histology analyses were performed 12 weeks postoperatively. In vitro data demonstrated that BM-MSCs, with MSC properties confirmed, remained vital after integration with ßTCP; and PLGA microspheres exhibited an initial burst followed by slow and continuous release of growth factors over a period of 28 days. In vivo data demonstrated that Group-B/-C sites had significantly greater gap obliteration, higher tissue mineral densities and more residual ßTCP granules (p<0.05, Kruskal-Wallis tests). Qualitatively, Group-B/-C defect sites had started remodeling while Group-A sites were mainly forming new bone to bridge the gaps. Furthermore, ßTCP degradation was not mediated by macrophages or osteoclasts, and was significantly slowed down by sealing the defects with barrier membrane. Combined, these data present a promising formulation composed of ßTCP granules, autologous MSCs, controlled-release growth factors and biodegradable PLGA barrier membrane for the reconstruction of critical-size mandibular defects.

4.
Mol Pharm ; 12(8): 2732-41, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26097994

ABSTRACT

The feasibility of various cellulose polymer derivatives, including methylcellulose (MC), hydroxypropyl methylcellulose (HPMC), sodium-carboxymethylcellulose (sodium-CMC), and cationic-hydroxyethylcellulose (cationic-HEC), for use as an excipient to enhance drug delivery in nasal spray formulations was investigated. Three main parameters for evaluating the polymers in nasal drug delivery applications include rheology, ciliary beat frequency (CBF), and permeation across nasal tissue. Reversible thermally induced viscosity enhancement was observed at near nasal physiological temperature when cellulose derivatives were combined with an additional excipient, poly(vinyl caprolactam)-poly(vinyl acetate)-poly(ethylene glycol) graft copolymer (PVCL-PVA-PEG). Cationic-HEC was shown to enhance acyclovir permeation across the nasal mucosa. None of the tested cellulosic polymers caused any adverse effects on porcine nasal tissues and cells, as assessed by alterations in CBF. Upon an increase in polymer concentration, a reduction in CBF was observed when ciliated cells were immersed in the polymer solution, and this decrease returned to baseline when the polymer was removed. While each cellulose derivative exhibited unique advantages for nasal drug delivery applications, none stood out on their own to improve more than one of the performance characteristics examined. Hence, these data may be useful for the development of new cellulose derivatives in nasal drug formulations.


Subject(s)
Cellulose/pharmacokinetics , Drug Carriers/pharmacokinetics , Nasal Mucosa/metabolism , Acyclovir/administration & dosage , Acyclovir/pharmacokinetics , Adhesiveness , Administration, Inhalation , Animals , Cells, Cultured , Cellulose/chemistry , Cilia/drug effects , Drug Carriers/chemistry , Drug Delivery Systems , Feasibility Studies , Nasal Mucosa/drug effects , Permeability , Polymers/chemistry , Polymers/pharmacokinetics , Rheology , Swine , Viscosity
5.
J Control Release ; 172(3): 662-70, 2013 Dec 28.
Article in English | MEDLINE | ID: mdl-24021356

ABSTRACT

An important poorly understood phenomenon in controlled-release depots involves the strong interaction between common cationic peptides and low Mw free acid end-group poly(lactic-co-glycolic acids) (PLGAs) used to achieve continuous peptide release kinetics. The kinetics of peptide sorption to PLGA was examined by incubating peptide solutions of 0.2-4mM octreotide or leuprolide acetate salts in a 0.1M HEPES buffer, pH7.4, with polymer particles or films at 4-37°C for 24h. The extent of absorption/loading of peptides in PLGA particles/films was assayed by two-phase extraction and amino acid analysis. Confocal Raman microspectroscopy, stimulated Raman scattering (SRS) and laser scanning confocal imaging, and microtome sectioning techniques were used to examine peptide penetration into the polymer phase. The release of sorbed peptide from leuprolide-PLGA particles was evaluated both in vitro (PBST+0.02% sodium azide, 37°C) and in vivo (male Sprague-Dawley rats). We found that when the PLGA-COOH chains are sufficiently mobilized, therapeutic peptides not only bind at the surface, a common belief to date, but also can be internalized and distributed throughout the polymer phase at physiological temperature forming a salt with low-molecular weight PLGA-COOH. Importantly, absorption of leuprolide into low MW PLGA-COOH particles yielded ~17 wt.% leuprolide loading in the polymer (i.e., ~70% of PLGA-COOH acids occupied), and the absorbed peptide was released from the polymer for >2 weeks in a controlled fashion in vitro and as indicated by sustained testosterone suppression in male Sprague-Dawley rats. This new approach, which bypasses the traditional encapsulation method and associated production cost, opens up the potential for facile production of low-cost controlled-release injectable depots for leuprolide and related peptides.


Subject(s)
Delayed-Action Preparations/chemistry , Lactic Acid/chemistry , Leuprolide/administration & dosage , Octreotide/administration & dosage , Polyglycolic Acid/chemistry , Adsorption , Animals , Drug Compounding , Leuprolide/chemistry , Male , Octreotide/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley
6.
Pharm Res ; 30(7): 1768-78, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23515830

ABSTRACT

PURPOSE: To investigate the effect of γ-irradiation of poly(lactic-co-glycolic acid) (PLGA)/Al(OH)3/0 or 5 wt% diethyl phthalate (DEP) microspheres for active self-healing encapsulation of vaccine antigens. METHODS: Microspheres were irradiated with 6°Co at 2.5 and 1.8 MRad and 0.37 and 0.20 MRad/h. Encapsulation of tetanus toxoid (TT) was achieved by mixing Al(OH)3-PLGA microspheres with TT solution at 10-38°C. Electron paramagnetic resonance (EPR) spectroscopy was used to examine free radical formation. Glass transition temperature (T(g)) and molecular weight of PLGA was measured by differential scanning calorimetry and gel permeation chromatography, respectively. Loading and release of TT were examined by modified Bradford, amino acid analysis, and ELISA assays. RESULTS: EPR spectroscopy results indicated absence of free radicals in PLGA microspheres after γ-irradiation. Antigen-sorbing capacity, encapsulation efficiency, and T(g) of the polymer were also not adversely affected. When DEP-loaded microspheres were irradiated at 0.2 MRad/h, some PLGA pores healed during irradiation and PLGA healing during encapsulation was suppressed. The molecular weight of PLGA was slightly reduced when DEP-loaded microspheres were irradiated at the same dose rate. At the 0.37 MRad/h dose rate, these trends were not observed and the full immunoreactivity of TT was preserved during encapsulation and 1-month release. Gamma irradiation slightly increased TT initial burst release. The small increase in total irradiation dose from 1.8 to 2.5 MRad had insignificant effect on the polymer and microspheres properties analyzed. CONCLUSIONS: Gamma irradiation is a plausible approach to provide a terminally sterilized, self-healing encapsulation PLGA excipient for vaccine delivery.


Subject(s)
Drug Delivery Systems , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , Sterilization , Tetanus Toxoid/administration & dosage , Gamma Rays , Microspheres , Polylactic Acid-Polyglycolic Acid Copolymer , Sterilization/methods , Transition Temperature
7.
J Control Release ; 165(1): 62-74, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23103983

ABSTRACT

Herein, we describe the detailed development of a simple and effective method to microencapsulate vaccine antigens in poly(lactic-co-glycolic acid) (PLGA) by simple mixing of preformed active self-microencapsulating (SM) PLGA microspheres in a low concentration aqueous antigen solution at modest temperature (10-38 °C). Co-encapsulating protein-sorbing vaccine adjuvants and polymer plasticizers were used to "actively" load the protein in the polymer pores and facilitate polymer self-healing at a temperature>the hydrated polymer glass transition temperature, respectively. The microsphere formulation parameters and loading conditions to provide optimal active self-healing microencapsulation of vaccine antigens in PLGA was investigated. Active self-healing encapsulation of two antigens, ovalbumin and tetanus toxoid (TT), in PLGA microspheres was adjusted by preparing blank microspheres containing different vaccine adjuvants (aluminum hydroxide (Al(OH)3) or calcium phosphate). Active loading of vaccine antigen in Al(OH)3-PLGA microspheres was found to: a) increase with an increasing loading of Al(OH)3 (0.88-3 wt.%) and addition of porosigen, b) decrease when the inner Al(OH)3/trehalose phase to 1 mL outer oil phase and size of microspheres was respectively >0.2 mL and 63 µm, and c) change negligibly by PLGA concentration and initial incubation (loading) temperature. Encapsulation of protein sorbing Al(OH)3 in PLGA microspheres resulted in suppression of self-healing of PLGA pores, which was then overcome by improving polymer chain mobility, which in turn was accomplished by coincorporating hydrophobic plasticizers in PLGA. Active self-healing microencapsulation of manufacturing process-labile TT in PLGA was found to: a) obviate micronization- and organic solvent-induced TT degradation, b) improve antigen loading (1.4-1.8 wt.% TT) and encapsulation efficiency (~97%), c) provide nearly homogeneous distribution and stabilization of antigen in polymer, and d) provide improved in vitro controlled release of antigenic TT.


Subject(s)
Antigens/chemistry , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , Tetanus Toxoid/chemistry , Vaccines , Adjuvants, Immunologic , Aluminum Hydroxide/chemistry , Calcium Phosphates/chemistry , Citrates/chemistry , Drug Compounding , Microspheres , Phthalic Acids/chemistry , Plasticizers/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Trehalose/chemistry
9.
Carcinogenesis ; 33(5): 1098-105, 2012 May.
Article in English | MEDLINE | ID: mdl-22427354

ABSTRACT

Systemic delivery of fenretinide in oral cancer chemoprevention trials has been largely unsuccessful due to dose-limiting toxicities and subtherapeutic intraoral drug levels. Local drug delivery, however, provides site-specific therapeutically relevant levels while minimizing systemic exposure. These studies evaluated the pharmacokinetic and growth-modulatory parameters of fenretinide mucoadhesive patch application on rabbit buccal mucosa. Fenretinide and blank-control patches were placed on right/left buccal mucosa, respectively, in eight rabbits (30 min, q.d., 10 days). No clinical or histological deleterious effects occurred. LC-MS/MS analyses of post-treatment samples revealed a delivery gradient with highest fenretinide levels achieved at the patch-mucosal interface (no metabolites), pharmacologically active levels in fenretinide-treated oral mucosa (mean: 5.65 µM; trace amounts of 4-oxo-4-HPR) and undetectable sera levels. Epithelial markers for cell proliferation (Ki-67), terminal differentiation (transglutaminase 1-TGase1) and glucuronidation (UDP-glucuronosyltransferase1A1-UGT1A1) exhibited fenretinide concentration-specific relationships (elevated TGase1 and UGT1A1 levels <5 µM, reduced Ki-67 indices >5 µM) relative to blank-treated epithelium. All fenretinide-treated tissues showed significantly increased intraepithelial apoptosis (TUNEL) positivity, implying activation of intersecting apoptotic and differentiation pathways. Human oral mucosal correlative studies showed substantial interdonor variations in levels of the enzyme (cytochrome P450 3A4-CYP3A4) responsible for conversion of fenretinide to its highly active metabolite, 4-oxo-4-HPR. Complementary in vitro assays in human oral keratinocytes revealed fenretinide and 4-oxo-4-HPR's preferential suppression of DNA synthesis in dysplastic as opposed to normal oral keratinocytes. Collectively, these data showed that mucoadhesive patch-mediated fenretinide delivery is a viable strategy to reintroduce a compound known to induce keratinocyte differentiation to human oral cancer chemoprevention trials.


Subject(s)
Fenretinide/administration & dosage , Mouth Neoplasms/prevention & control , Animals , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Chemoprevention/methods , Cytochrome P-450 CYP3A/metabolism , Drug Delivery Systems/methods , Epithelium/drug effects , Epithelium/metabolism , Female , Fenretinide/analogs & derivatives , Fenretinide/metabolism , Fenretinide/pharmacokinetics , Glucuronosyltransferase/metabolism , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Mouth Mucosa/drug effects , Mouth Mucosa/metabolism , Mouth Neoplasms/metabolism , Rabbits
10.
Mol Pharm ; 9(4): 937-45, 2012 Apr 02.
Article in English | MEDLINE | ID: mdl-22280430

ABSTRACT

The objective of this study was to enhance oral mucosal permeation of fenretinide by coincorporation of propylene glycol (PG) and menthol in fenretinide/Eudragit RL PO mucoadhesive patches. Fenretinide is an extremely hydrophobic chemopreventive compound with poor tissue permeability. Coincorporation of 5-10 wt % PG (mean J(s) = 16-23 µg cm⁻² h⁻¹; 158-171 µg of fenretinide/g of tissue) or 1-10 wt % PG + 5 wt % menthol (mean J(s) = 18-40 µg cm⁻² h⁻¹; 172-241 µg of fenretinide/g of tissue) in fenretinide/Eudragit RL PO patches led to significant ex vivo fenretinide permeation enhancement (p < 0.001). Addition of PG above 2.5 wt % in the patch resulted in significant cellular swelling in the buccal mucosal tissues. These alterations were ameliorated by combining both enhancers and reducing PG level. After buccal administration of patches in rabbits, in vivo permeation of fenretinide across the oral mucosa was greater (∼43 µg fenretinide/g tissue) from patches that contained optimized permeation enhancer content (2.5 wt % PG + 5 wt % menthol) relative to permeation obtained from enhancer-free patch (∼17 µg fenretinide/g tissue) (p < 0.001). In vitro and in vivo release of fenretinide from patch was not significantly increased by coincorporation of permeation enhancers, indicating that mass transfer across the tissue, and not the patch, largely determined the permeation rate control in vivo. As a result of its improved permeation and its lack of deleterious local effects, the mucoadhesive fenretinide patch coincorporated with 2.5 wt % PG + 5 wt % menthol represents an important step in the further preclinical evaluation of oral site-specific chemoprevention strategies with fenretinide.


Subject(s)
Fenretinide/pharmacokinetics , Menthol/chemistry , Mouth Mucosa/metabolism , Mouth Neoplasms/prevention & control , Propylene Glycol/chemistry , Animals , Chemoprevention/methods , In Vitro Techniques , Rabbits , Solubility , Swine
11.
J Carcinog ; 10: 23, 2011.
Article in English | MEDLINE | ID: mdl-22013393

ABSTRACT

Due to its characterized progression from recognized premalignant oral epithelial changes (i.e., oral epithelial dysplasia) to invasive cancer, oral squamous cell carcinoma represents an optimal disease for chemopreventive intervention prior to malignant transformation. The primary goal of oral cancer chemoprevention is to reverse, suppress, or inhibit the progression of premalignant lesions to cancer. Over the last several decades, numerous oral cancer chemoprevention clinical trials have assessed the therapeutic efficacy of diverse chemopreventive agents. The standard of care for more advanced oral dysplastic lesions entails surgical excision and close clinical follow-up due to the potential (~33%) for local recurrence at a similar or more advanced histological stage. The purpose of this review was to identify prominent oral cancer chemoprevention clinical trials, assess their overall therapeutic efficacy, and delineate effects of local versus systemic drug administration. In addition, these compiled clinical trial data present concepts for consideration in the design and conduction of future clinical trials.

12.
Pharm Res ; 28(10): 2599-609, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21674264

ABSTRACT

PURPOSE: To develop fenretinide oral mucoadhesive patch formulations and evaluate their in vitro and in vivo release performance for future site-specific chemoprevention of oral cancer. METHODS: Solubilization of fenretinide in simulated saliva (SS) was studied by incorporating nonionic surfactants (Tween® 20 and 80, and Brij® 35 and 98), bile salts (sodium salt of cholic, taurocholic, glycocholic, and deoxycholic acids), phospholipid (lecithin), and novel polymeric solubilizer (Souplus®). Adhesive (polycarbophil: hydroxypropyl methylcellulose 4KM) and drug release (Fenretinide/Eudragit® RL PO with or without solubilizers) layers were prepared by solvent casting. Oral mucoadhesive patches were formed by attaching drug and adhesive layers onto backing layer (Tegaderm™ film). Physical state of drug in Eudragit® films was examined by X-ray diffraction (XRD). Evaluation of in vitro and in vivo fenretinide release from the patch was conducted in SS containing 5%w/v sodium deoxycholate and rabbits, respectively. Fenretinide was quantified by HPLC. RESULTS: Tween® 20 and 80, Brij® 98, and sodium deoxycholate exhibited the highest fenretinide solubilization potential among the solubilizers. Drug loading efficiency in Eudragit® films was 90%-97%. XRD suggested fenretinide was amorphous in solubilizer-free and solubilizer-loaded films. Solubilizer-free patch exhibited poor in vitro and in vivo controlled drug release behavior. Increases in drug loading (5-10 wt%) or changes in polymeric matrix permeability did not provide continuous drug release. Co-incorporation of either single or mixed solubilizers in fenretinide/Eudragit® patches, (20 wt% Tween® 20, Tween® 80 and sodium deoxycholate or 20 wt% Tween® 80 + 40 wt% sodium deoxycholate solubilizers) led to significantly improved continuous in vitro/in vivo fenretinide release. CONCLUSION: Fenretinide/Eudragit® RL PO patches with 20 wt% Tween® 80 + 40 wt% sodium deoxycholate solubilizers exhibit excellent release behavior for further preclinical and/or clinical evaluation in oral cancer chemoprevention.


Subject(s)
Fenretinide/administration & dosage , Fenretinide/chemistry , Mouth Mucosa/metabolism , Mouth Neoplasms/drug therapy , Adhesives/chemistry , Administration, Buccal , Animals , Bile Acids and Salts/chemistry , Chemoprevention/methods , Dosage Forms , Drug Design , Female , Mouth Neoplasms/metabolism , Phospholipids/chemistry , Polymers/chemistry , Rabbits , Saliva/drug effects , Solubility , Surface-Active Agents/chemistry
13.
Pharm Res ; 27(4): 628-43, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20148292

ABSTRACT

PURPOSE: The objective of this study was to formulate and evaluate freeze-dried black raspberry (FBR) ethanol extract (RE) loaded poly(DL-lactic-co-glycolic acid) (PLGA) and poly(DL-lactic acid) (PLA) injectable millicylindrical implants for sustained delivery of chemopreventive FBR anthocyanins (cyanidin-3-sambubioside (CS), cyanidin-3-glucoside (CG) and cyanidin-3-rutinoside (CR)). METHODS: Identification and quantitation of CS, CG, and CR in RE was performed by mass spectroscopy and HPLC. RE:triacetyl-beta-cyclodextrin (TA-beta-CD) inclusion complex (IC) was prepared by a kneading method and characterized by X-ray diffraction (XRD), nuclear magnetic resonance spectroscopy (NMR) and UV-visible spectroscopy. RE or RE:TA-beta-CD IC-loaded PLGA or PLA implants were prepared by a solvent extrusion method. In vitro and in vivo controlled release studies were conducted in phosphate-buffered saline Tween-80 (pH 7.4, 37 degrees C) and after subcutaneous administration in male Sprague-Dawley rats, respectively. Anthocyanins were quantified by HPLC at 520 nm. RESULTS: The content of CS, CG, and CR in RE was 0.2, 1.5, and 3.5 wt%, respectively. The chemical stability of anthocyanins in solution was determined to be pH-dependent, and their degradation rate increased with an increase in pH from 2.4 to 7.4. PLGA/PLA millicylindrical implants loaded with 5 or 10 wt% RE exhibited a high initial burst and short release duration of anthocyanins (35-52 and 80-100% CG + CR release after 1 and 14 days, respectively). The cause for rapid anthocyanins release was linked to higher polymer water uptake and porosity associated with the high osmolytic components of large non-anthocyanin fraction of RE. XRD, (1)H NMR and UV-visible spectroscopy indicated that the non-anthocyanin fraction molecules of RE formed an IC with TA-beta-CD, decreasing the hydrophilicity of RE. Formation of an IC with hydrophobic carrier, TA-beta-CD, provided better in vitro/in vivo sustained release of FBR anthocyanins (16-24 and 97-99% CG + CR release, respectively, after 1 and 28 days from 20 wt% RE:TA-beta-CD IC/PLA implants) over 1 month, owing to reduced polymer water uptake and porosity. CONCLUSION: PLA injectable millicylindrical implants loaded with RE:TA-beta-CD IC are optimal dosage forms for 1-month slow and continuous delivery of chemopreventive FBR anthocyanins.


Subject(s)
Anthocyanins/administration & dosage , Anthocyanins/therapeutic use , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Delayed-Action Preparations/chemistry , Fruit/chemistry , Rosaceae/chemistry , Animals , Anthocyanins/analysis , Anthocyanins/pharmacology , Antineoplastic Agents, Phytogenic/analysis , Antineoplastic Agents, Phytogenic/pharmacology , Injections , Lactic Acid/chemistry , Male , Polyesters , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers/chemistry , Prostheses and Implants , Rats , Rats, Sprague-Dawley
14.
Eur J Pharm Biopharm ; 70(1): 187-98, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18472254

ABSTRACT

The objective of this study was to investigate the potential of various formulation strategies to achieve 1-month continuous (improved) release of the novel anti-cancer drug, 2-methoxyestradiol (2-ME), from injectable cylindrical poly(DL-lactide-co-glycolide) (PLGA) implants. PLGA implants were prepared by a solvent extrusion method. PLGA 50:50 (M(w)=51 kDa, end group=lauryl ester) (PLGA-lauryl ester) implants loaded with 3-30 wt% 2-ME exhibited a pronounced lag phase (i.e., corresponding to induction time to polymer mass loss) and triphasic release profile. Incorporation of 5 wt% hydroxypropyl-beta-cyclodextrin (HP-beta-CD) (approximately 57% release after 28 days) or Pluronic F127 (approximately 42% release after 28 days) in PLGA-lauryl ester implants reduced the lag-phase and improved the drug release moderately over a period of 28 days. The formation and the incorporation of a 2-ME/polyethylene glycol (PEG) 8000 solid dispersion in PLGA-lauryl ester implants further increased drug release (approximately 21% and 73% release after 1 and 28 days, respectively), attributable to improved drug solubility/dissolution, higher matrix porosity, and accelerated polymer degradation. Blending of PLGA 50:50 (M(w)=24 kDa, end group=COOH) (PLGA-COOH) with the PLGA-lauryl ester also provided moderate enhancement of 2-ME release over a period of 28 days. PLGA-COOH (M(w)=24 kDa) implants with 3-5% w/w pore-forming MgCO(3) exhibited the most desirable drug release among all the formulations tested, and, demonstrated 1-month slow and continuous in vitro release of approximately 80% 2-ME after a minimal initial burst. Hence, these formulation approaches provide several possible avenues to improve release rates of the hydrophobic drug, 2-ME, from PLGA for future application in regional anti-cancer therapy.


Subject(s)
Angiogenesis Inhibitors/chemistry , Drug Carriers , Drug Implants , Estradiol/analogs & derivatives , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , 2-Hydroxypropyl-beta-cyclodextrin , 2-Methoxyestradiol , Angiogenesis Inhibitors/administration & dosage , Chemistry, Pharmaceutical , Drug Compounding , Estradiol/administration & dosage , Estradiol/chemistry , Excipients/chemistry , Injections , Kinetics , Lauric Acids/chemistry , Magnesium/chemistry , Molecular Weight , Poloxamer/chemistry , Polyethylene Glycols/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Porosity , Solubility , Technology, Pharmaceutical/methods , Water/chemistry , beta-Cyclodextrins/chemistry
15.
Pharm Res ; 25(3): 586-97, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17891553

ABSTRACT

PURPOSE: The objective of this study was to develop poly(lactic-co-glycolic acid) (PLGA) injectable implants (i.e., millicylinders) with microencapsulated N-acetylcysteine (NAC) for site-specific controlled NAC release, for potential chemopreventive applications in persons with previously excised head and neck cancers. METHODS: PLGA 50:50 (i.v.=0.57 dl/g) implants with 1-10 wt% NAC free acid or 10 wt% NAC salts (NAC-Na+, NAC-Mg2+ and NAC-Ca2+) were prepared by solvent extrusion and/or fluid energy micronization (FEM) methods. X-ray diffraction (XRD), scanning electron microscopy (SEM), and differential scanning calorimetry (DSC) studies were performed to evaluate the physical mixing of NAC with PLGA. PLGA implant degradation was studied by kinetics of polymer molecular weight decline (gel permeation chromatography) and mass loss. Release studies were conducted in N2 purged PBS (pH 7.4) at 37 degrees C in evacuated and sealed ampoules. NAC was quantified by HPLC at 210 nm. RESULTS: XRD, SEM and DSC studies indicated that NAC had dissolved in the polymer phase at 1-3.5% w/w loading, but became discretely suspended in the polymer at 6-10% w/w. Initial burst and long-term release rate increased with increased drug loading, and release was uncharacteristically rapid at higher loading (6-10% w/w). The cause of the rapid release was linked to extensive plasticization, matrix porosity and general acid catalysis of PLGA degradation caused by the NAC free acid. PLGA millicylinders loaded with 10% w/w NAC-Ca2+ and NAC-Mg2+salts exhibited reduced burst (34 vs 13-22% release within a day of incubation for NAC free acid vs NAC-Ca2+ and NAC-Mg2+salts, respectively) and slow and continuous complete release over 4 weeks without significant NAC-catalyzed degradation of PLGA. Release of NAC from NAC-Ca2+/PLGA implant was slower than that of NAC-Mg2+/PLGA consistent with the lower solubility of the former salt. NAC with its free thiol was rapidly converted to its cystine dimer in the presence of molecular oxygen. PLGA released samples in sealed and evacuated ampoules indicated>80% parent NAC remaining after the 1 month release analysis irrespective of initial NAC free acid and salt forms. CONCLUSION: By encapsulating the NAC-Mg2+ and NAC-Ca2+ salts in PLGA implants, the high initial burst, short release duration, and the general acid catalysis caused by the NAC free acid were each prevented and 1-month slow and continuous release was attained with minimal instability of the free thiol group.


Subject(s)
Acetylcysteine/chemistry , Antineoplastic Agents/chemistry , Drug Carriers , Lactic Acid/chemistry , Matrix Metalloproteinase Inhibitors , Polyglycolic Acid/chemistry , Polymers/chemistry , Protease Inhibitors/chemistry , Acetylcysteine/administration & dosage , Acetylcysteine/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Calcium Compounds/chemistry , Calorimetry, Differential Scanning , Catalysis , Chemistry, Pharmaceutical , Drug Compounding , Drug Implants , Drug Stability , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/surgery , Humans , Injections , Kinetics , Magnesium Compounds/chemistry , Microscopy, Electron, Scanning , Polylactic Acid-Polyglycolic Acid Copolymer , Protease Inhibitors/administration & dosage , Protease Inhibitors/pharmacology , Solubility , Technology, Pharmaceutical , X-Ray Diffraction
17.
J Agric Food Chem ; 53(5): 1728-33, 2005 Mar 09.
Article in English | MEDLINE | ID: mdl-15740066

ABSTRACT

Trypsin was immobilized on linolenic acid modified chitosan using glutaraldehyde (GA) as cross-linker, which was confirmed by Fourier transform infrared (FTIR) spectra. The chitosan nanoparticles containing trypsin (TR) can be prepared after the sonication of immobilized trypsin. The GA concentration affected both the enzyme activity of the nanoparticle and particle size. Results indicated that the activity of trypsin immobilized onto linolenic acid modified chitosan nanoparticles increased with increasing concentration of GA up to 0.07% (v/v) and then decreased with increasing amount of GA. On the other hand, particle size increased (from 523 to 1372 nm) with the increasing concentration of GA (from 0.03 to 0.1% v/v). The enzyme catalytic characteristics of nanoparticle solution were also studied. The results showed that the kinetic constant value (K(m)) of TR immobilized on nanoparticle (71.9 mg/mL) was higher than that of pure TR (50.2 mg/mL). However, the thermal stability and optimum temperature of TR immobilized on nanoparticles improved, which make it more attractive in the application aspect.


Subject(s)
Chitosan/chemistry , Nanostructures/chemistry , Trypsin/analysis , Catalysis , Cross-Linking Reagents , Enzymes, Immobilized , Glutaral/analysis , Glutaral/chemistry , Kinetics , Particle Size , Solutions , Spectroscopy, Fourier Transform Infrared , Trypsin/metabolism , alpha-Linolenic Acid/chemistry
18.
Drug Dev Ind Pharm ; 31(1): 1-10, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15704852

ABSTRACT

The aim of the present study was to enhance the dissolution rate of valdecoxib using its solid dispersions (SDs) with polyethylene glycol (PEG) 4000. The phase solubility behavior of valdecoxib in the presence of various concentrations of PEG 4000 in water was obtained at 37 degrees C. The solubility of valdecoxib increased with increasing amount of PEG 4000 in water. Gibbs free energy (deltaG(zero)tr) values were all negative, indicating the spontaneous nature of valdecoxib solubilization, and they decreased with increase in the PEG 4000 concentration, demonstrating that the reaction conditions became more favorable as the concentration of PEG 4000 increased. The SDs of valdecoxib with PEG 4000 were prepared at 1:1, 1:2, 1:5, and 1:10 (valdecoxib: PEG 4000) ratio by melting method. Evaluation of the properties of the SDs was performed by using dissolution, Fourier-transform infrared (FTIR) spectroscopy, differential scanning calorimetry (DSC), X-ray diffraction (XRD), and scanning electron microscopy (SEM) studies. The SDs of valdecoxib with PEG 4000 exhibited enhanced dissolution rate of valdecoxib, and the rate increased with increasing concentration of PEG 4000 in SDs. Mean dissolution time (MDT) of valdecoxib decreased significantly after preparation of SDs and physical mixture with PEG 4000. The FTIR spectroscopic studies showed the stability of valdecoxib and absence of well-defined valdecoxib-PEG 4000 interaction. The DSC and XRD studies indicated the amorphous state of valdecoxib in SDs of valdecoxib with PEG 4000. The SEM pictures showed the formation of effective SDs of valdecoxib with PEG 4000, since well-defined changes in the surface nature of valdecoxib, SDs, and physical mixture were observed.


Subject(s)
Cyclooxygenase Inhibitors/chemistry , Isoxazoles/chemistry , Polyethylene Glycols , Sulfonamides/chemistry , Algorithms , Calorimetry, Differential Scanning , Chemistry, Pharmaceutical , Excipients , Kinetics , Mannitol , Microscopy, Electron, Scanning , Povidone , Solubility , Spectroscopy, Fourier Transform Infrared , Urea , X-Ray Diffraction
19.
J Agric Food Chem ; 53(2): 437-41, 2005 Jan 26.
Article in English | MEDLINE | ID: mdl-15656685

ABSTRACT

Chitosan was modified by coupling with linolenic acid through the 1-ethyl-3-(3-dimethylaminopropyyl)carbodiimide-mediated reaction. The degree of substitution was measured by 1H NMR, and it was 1.8%, i.e., 1.8 linolenic acids group per 100 anhydroglucose units. The critical aggregation concentration (CAC) of the self-aggregate of hydrophobically modified chitosan was determined by measuring the fluorescence intensity of the pyrene as a fluorescent probe. The CAC value in phosphate-buffered saline (PBS) solution (pH 7.4) was 5 x 10(-2) mg/mL. The average particle size of self-aggregates of hydrophobically modified chitosan in PBS solution (pH 7.4) was 210.8 nm with a unimodal size distribution ranging from 100 to 500 nm. A transmission electron microscopy study showed that the formation of near spherical shape nanoparticles had enough structural integrity. The loading ability of hydrophobically modified chitosan (LA-chitosan) was investigated by using bovine serum albumin (BSA) as a model protein. Self-aggregated nanoparticles exhibited an increased loading capacity (19.85 +/- 0.04 to 37.57 +/- 0.25%) with an increasing concentration of BSA (0.1-0.5 mg/mL).


Subject(s)
Chitosan/chemistry , Nanostructures , alpha-Linolenic Acid/chemistry , Fluorescent Dyes , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Microscopy, Electron , Particle Size , Serum Albumin, Bovine/chemistry , Spectrometry, Fluorescence , Spectroscopy, Fourier Transform Infrared
SELECTION OF CITATIONS
SEARCH DETAIL
...