Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Publication year range
1.
Biotechnol Bioeng ; 118(7): 2804-2814, 2021 07.
Article in English | MEDLINE | ID: mdl-33913523

ABSTRACT

The application of scaffold-based stem cell transplantation to enhance peripheral nerve regeneration has great potential. Recently, the neuroregenerative potential of tacrolimus (a U.S. Food and Drug Administration-approved immunosuppressant) has been explored. In this study, a fibrin gel-based drug delivery system for sustained and localized tacrolimus release was combined with rat adipose-derived mesenchymal stem cells (MSC) to investigate cell viability in vitro. Tacrolimus was encapsulated in poly(lactic-co-glycolic) acid (PLGA) microspheres and suspended in fibrin hydrogel, using concentrations of 0.01 and 100 ng/ml. Drug release over time was measured. MSCs were cultured in drug-released media collected at various days to mimic systemic exposure. MSCs were combined with (i) hydrogel only, (ii) empty PLGA microspheres in the hydrogel, (iii) 0.01, and (iv) 100 ng/ml of tacrolimus PLGA microspheres in the hydrogel. Stem cell presence and viability were evaluated. A sustained release of 100 ng/ml tacrolimus microspheres was observed for up to 35 days. Stem cell presence was confirmed and cell viability was observed up to 7 days, with no significant differences between groups. This study suggests that combined delivery of 100 ng/ml tacrolimus and MSCs in fibrin hydrogel does not result in cytotoxic effects and could be used to enhance peripheral nerve regeneration.


Subject(s)
Drug Delivery Systems , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Nerve Regeneration , Peripheral Nerve Injuries , Animals , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/therapy , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Rats , Tacrolimus/chemistry , Tacrolimus/pharmacokinetics , Tacrolimus/pharmacology
2.
Cell Biol Int ; 45(8): 1733-1742, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33851759

ABSTRACT

The nerve conduits have been developed for nerve defect repair. However, no artificial conduits have obtained comparable results to autografts to bridge the large gaps. A possible reason for this poor performance may be a lack of sustainable neurotrophic support for axonal regrowth. Previous studies suggested nanocomposite conduits can be used as a carrier for valproic acid (VPA), a common drug that can produce effects similar to the neurotrophic factors. Here, we developed the novel bioabsorbable conduits based on hydroxyapatite/poly d-l-lactic acid (PDLLA)/poly{(lactic acid)-co-[(glycolic acid)-alt-(l-lysine)]} with sustained release of VPA. Firstly, the sustained release of VPA in this conduit was examined by high-performance liquid chromatography. Then Schwann cells were treated with the conduit extracts. The cell metabolic activity and proliferation were assayed by 3-[4,5-dimethyl-2-thiazolyl]-2,5-diphenyl-2-tetrazolium bromide and bromodeoxyuridine staining. A 10-mm segment of rat sciatic nerve was resected and then repaired, respectively, using the VPA conduit (Group A), the PDLLA conduit (Group B), or the autografts (Group C). Nerve conduction velocities (NCVs), compound muscle action potentials (CMAPs), and histological staining were assayed following the surgery. The cell metabolic activity and proliferation were significantly increased (p < .05) by the extracts from VPA-conduit extract compared to others. NCVs and CMAPs were significantly higher in Groups A and C than Group B (p < .05). The nerve density of Groups A and C was higher than Group B. There was no significant difference between Groups A and C. Taken together, this study suggested the sustained-release VPA conduit promoted peripheral nerve regeneration that was comparable to the autografts. It holds potential for future use in nerve regeneration.


Subject(s)
Biocompatible Materials/pharmacokinetics , Durapatite/pharmacokinetics , Nerve Regeneration/drug effects , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Sciatic Nerve/drug effects , Valproic Acid/pharmacokinetics , Animals , Animals, Newborn , Biocompatible Materials/administration & dosage , Cells, Cultured , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Liberation/drug effects , Drug Liberation/physiology , Durapatite/administration & dosage , Male , Nerve Regeneration/physiology , Polyesters/administration & dosage , Polyesters/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Rats , Rats, Wistar , Schwann Cells/drug effects , Schwann Cells/metabolism , Sciatic Nerve/metabolism , Valproic Acid/administration & dosage
3.
Acta Biochim Biophys Sin (Shanghai) ; 53(8): 1027-1036, 2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34109980

ABSTRACT

Targeted delivery and smart response of nanomedicine hold great promise for improving the therapeutic efficacy and alleviating the side effects of chemotherapy agents in cancer treatment. However, availability of only a few studies that discuss organic nanomedicines with these properties limits the development prospects of nanomedicines. In the present study, folic acid (FA)-targeted delivery and glutathione (GSH) smart responsive nanomedicine were rationally designed for paclitaxel (PTX) delivery for the treatment of lung cancer. Compared with other stimuli-responsive nanomedicines, this nanocarrier was not only sensitive to biologically relevant GSH for on-demand drug release but also biodegradable into biocompatible products after fulfilling its delivery task. The nanomedicine first entered tumor cells via FA and its receptor-mediated endocytosis. After the lysosomal escape, poly(lactic-co-glycolic acid) (PLGA) nanomedicine was triggered by a higher level of GSH and released its cargo into the tumor microenvironment. In vitro and in vivo results revealed that the PLGA nanomedicine not only inhibited the proliferation and promoted the apoptosis of lung cancer cells significantly but also possessed less toxic side effects when compared with free PTX. Therefore, the proposed drug delivery system demonstrates the potential of a multifunctional nano-platform to enhance bioavailability and reduce the side effects of chemotherapy agents.


Subject(s)
Carcinoma, Lewis Lung , Folic Acid , Glutathione/metabolism , Lung Neoplasms , Nanomedicine , Paclitaxel , Polylactic Acid-Polyglycolic Acid Copolymer , Animals , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Folic Acid/chemistry , Folic Acid/pharmacokinetics , Folic Acid/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology
4.
Int J Mol Sci ; 22(6)2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33801871

ABSTRACT

Recently, nano- and micro-particulate systems have been widely utilized to deliver pharmaceutical compounds to achieve enhanced therapeutic effects and reduced side effects. Poly (DL-lactide-co-glycolide) (PLGA), as one of the biodegradable polyesters, has been widely used to fabricate particulate systems because of advantages including controlled and sustained release, biodegradability, and biocompatibility. However, PLGA is known for low encapsulation efficiency (%) and insufficient controlled release of water-soluble drugs. It would result in fluctuation in the plasma levels and unexpected side effects of drugs. Therefore, the purpose of this work was to develop microcapsules loaded with alginate-coated chitosan that can increase the encapsulation efficiency of the hydrophilic drug while exhibiting a controlled and sustained release profile with reduced initial burst release. The encapsulation of nanoparticles in PLGA microcapsules was done by the emulsion solvent evaporation method. The encapsulation of nanoparticles in PLGA microcapsules was confirmed by scanning electron microscopy and confocal microscopy. The release profile of hydrophilic drugs can further be altered by the chitosan coating. The chitosan coating onto alginate exhibited a less initial burst release and sustained release of the hydrophilic drug. In addition, the encapsulation of alginate nanoparticles and alginate nanoparticles coated with chitosan in PLGA microcapsules was shown to enhance the encapsulation efficiency of a hydrophilic drug. Based on the results, this delivery system could be a promising platform for the high encapsulation efficiency and sustained release with reduced initial burst release of the hydrophilic drug.


Subject(s)
Delayed-Action Preparations/pharmacokinetics , Nanoparticles/chemistry , Pharmaceutical Preparations/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Alginates/chemistry , Biodegradation, Environmental , Capsules , Chitosan/chemistry , Delayed-Action Preparations/chemistry , Drug Carriers , Drug Liberation , Hydrophobic and Hydrophilic Interactions , Microscopy, Electron, Scanning , Microspheres , Nanoparticles/ultrastructure , Particle Size , Pharmaceutical Preparations/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
5.
Mol Pharm ; 17(2): 441-452, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31886676

ABSTRACT

Drug delivery by direct intraductal administration can achieve high local drug concentration in the breast and minimize systemic levels. However, the clinical application of this approach for breast cancer treatment is limited by the rapid clearance of the drug from the ducts. With the goal of developing strategies to prolong drug retention in the breast, this study was focused on understanding the influence of particle size and formulation on breast duct and lymph node retention. Fluorescent-labeled polystyrene (PS) particles ranging in size from 100 to 1000 nm were used to study the influence of particle size. Polylactic acid-co-glycolic acid (PLGA) was used to develop and test formulations for intraductal delivery. Cy 5.5, a near-IR dye, was encapsulated in PLGA microparticles, nanoparticles, and the in situ gel to study the biodistribution in rats using an in vivo imager. PS microparticles (1 µm) showed longer retention in the duct compared to 100 and 500 nm nanoparticles. The ductal retention half-life was 5-fold higher for PS microparticles compared to the nanoparticles. On the other hand, the free dye was cleared from the breast within 6 h. PLGA nanoparticles sustained the release of Cy 5.5 for >4 days. Microparticles and gel showed a much slower release than nanoparticles. PLGA in situ gel and microparticles were retained in the breast for up to 4 days, while the nanoparticles were retained in the breast for 2 days. PLGA nanoparticles and microparticles drained to the axillary lymph node and were retained for up to 24 and 48 h, respectively, while the in situ gel and the free dye did not show any detectable fluorescence in the lymph nodes. Taken together, the results demonstrate the feasibility of prolonged retention in the breast duct and lymph node by optimal formulation design. The findings can serve as a framework to design formulations for localized treatment of breast cancer.


Subject(s)
Drug Compounding , Drug Delivery Systems/methods , Lymph Nodes/drug effects , Mammary Glands, Animal/drug effects , Nanoparticles/administration & dosage , Particle Size , Animals , Breast Neoplasms/drug therapy , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Drug Liberation , Female , Half-Life , Lymph Nodes/metabolism , Mammary Glands, Animal/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polystyrenes/chemistry , Polystyrenes/pharmacokinetics , Rats , Rats, Sprague-Dawley , Tissue Distribution
6.
J Thromb Thrombolysis ; 49(3): 404-412, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31898270

ABSTRACT

The purpose of the currents study was to enhance bioavailability of rivaroxaban (RXB) and reduce the food effect. RXB loaded PLGA nanoparticles (RXB-PLGA-NPs) were prepared by emulsion solvent evaporation method and optimized using central composite design (CDD). The optimized RXB-PLGA-NPs (F8) with composition, PLGA (125 mg), PVA (0.5%w/w) and RXB (20 mg) was found optimum with particle size (496 ± 8.5 nm), PDI (0.607), ZP (- 18.41 ± 3.14 mV), %EE (87.9 ± 8.6) and %DL (9.5 ± 1.6). The optimized NPs (F8) was further evaluated in vitro for DSC, FTIR, SEM and in vitro release studies. A comparative pharmacokinetic studies with commercial tablet (XARELTO®) were conducted on fasted and fed state rats. Compared to commercial tablet (XARELTO®), the RXB-PLGA-NPs (F8) exhibited a significant enhancement of bioavailability in both fasted and fed state. In addition, the bioavailability of RXB from NPs (F8) was found unaffected in the presence of food.


Subject(s)
Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer , Rivaroxaban , Administration, Oral , Animals , Biological Availability , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Food-Drug Interactions , Male , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Rats , Rats, Wistar , Rivaroxaban/chemistry , Rivaroxaban/pharmacokinetics , Rivaroxaban/pharmacology
7.
Nanomedicine ; 30: 102291, 2020 11.
Article in English | MEDLINE | ID: mdl-32841737

ABSTRACT

Polylactide-co-glycolide (PLGA) nanoparticles are one of the most commonly explored biodegradable polymeric drug carriers for inhaled delivery. Despite their advantages as inhalable nanomedicine scaffolds, we still lack a complete understanding of the kinetics and major pathways by which these materials are cleared from the lungs. This information is important to evaluate their safety over prolonged use and enable successful clinical translation. This study aimed to determine how the size and charge of 3H-labeled PLGA nanoparticles affect the kinetics and mechanisms by which they are cleared from the lungs and their safety in the lungs. The results showed that lung clearance kinetics and retention patterns were more significantly defined by particle size, whereas lung clearance pathways were largely influenced by particle charge. Each of the nanoparticles caused transient inflammatory changes in the lungs after a single dose that reflected lung retention times.


Subject(s)
Lung/metabolism , Nanoparticles/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Animals , Bronchoalveolar Lavage Fluid , Drug Administration Routes , Lung/immunology , Male , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/blood , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Rats , Rats, Sprague-Dawley , Tissue Distribution , Trachea
8.
J Mater Sci Mater Med ; 31(12): 115, 2020 Nov 28.
Article in English | MEDLINE | ID: mdl-33247423

ABSTRACT

Persistent local oxygen delivery is crucial to create a microenvironment for cell survival and nerve regeneration in acute spinal cord injury (SCI). This study aimed to fabricate calcium peroxide-based microspheres incorporated into a 3-D construct scaffold as a novel oxygen release therapy for SCI. The scaffolds were able to generate oxygen over the course of 21 days when incubated under hypoxic conditions. In vitro, GFP-labeled bone marrow-derived mesenchymal stem cells (MSCs) were planted into the scaffolds. We observed that scaffolds could enhance MSC survival under hypoxic conditions for more than 21 days. Oxygen generating scaffolds were transplanted into spinal cord injury sites of rats in vivo. Twelve weeks following transplantation, cavity areas in the injury/graft site were significantly reduced due to tissue regeneration. Additionally, the oxygen generating scaffolds improved revascularization as observed through vWF immunostaining. A striking feature was the occurrence of nerve fiber regeneration in the lesion sites, which eventually led to significant locomotion recovery. The present results indicate that the oxygen generating scaffolds have the property of sustained local oxygen release, thus facilitating regeneration in injured spinal cords.


Subject(s)
Coated Materials, Biocompatible , Guided Tissue Regeneration , Oxygen/pharmacokinetics , Spinal Cord Injuries/rehabilitation , Tissue Scaffolds , Animals , Cell Survival/drug effects , Cells, Cultured , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacokinetics , Coated Materials, Biocompatible/pharmacology , Female , Guided Tissue Regeneration/instrumentation , Guided Tissue Regeneration/methods , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Microspheres , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Oxygen/metabolism , Oxygen/pharmacology , Peroxides/chemistry , Peroxides/pharmacokinetics , Peroxides/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/therapy , Tissue Scaffolds/chemistry
9.
Nano Lett ; 19(8): 5587-5594, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31260628

ABSTRACT

In breast cancer chemophotothermal therapy, it is a great challenge for the development of multifunctional nanoagents for precision targeting and the effective treatment of tumors, especially for metastasis. Herein, we successfully design and synthesize a multifunctional black phosphorus (BP)-based nanoagent, BP/DTX@PLGA, to address this challenge. In this composite nanoagent, BP quantum dots (BPQDs) are loaded into poly(lactic-co-glycolic acid) (PLGA) with additional conjugation of a chemotherapeutic agent, docetaxel (DTX). The in vivo distribution results demonstrate that BP/DTX@PLGA shows striking tropism for targeting both primary tumors and lung metastatic tumors. Moreover, BP/DTX@PLGA exhibits outstanding controllable chemophotothermal combinatory therapeutics, which dramatically improves the efficacy of photothermal tumor ablation when combined with near-light irradiation. Mechanistically, accelerated DTX release from the nanocomplex upon heating and thermal treatment per se synergistically incurs apoptosis-dependent cell death, resulting in the elimination of lung metastasis. Meanwhile, in vitro and in vivo results further confirm that BP/DTX@PLGA possesses good biocompatibility. This study provides a promising BP-based multimodal nanoagent to constrain cancer metastasis.


Subject(s)
Antineoplastic Agents/therapeutic use , Docetaxel/therapeutic use , Mammary Neoplasms, Animal/therapy , Nanoconjugates/therapeutic use , Phosphorus/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Docetaxel/pharmacokinetics , Female , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mammary Neoplasms, Animal/pathology , Mice , Neoplasm Metastasis/pathology , Neoplasm Metastasis/therapy , Phosphorus/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/therapeutic use
10.
Biotechnol Bioeng ; 116(7): 1777-1794, 2019 07.
Article in English | MEDLINE | ID: mdl-30905072

ABSTRACT

Poly-lactic-co-glycolic acid (PLGA) microcarriers (0.8 ± 0.2 µm) have been fabricated with a load of 20 µg/gPLGA by an emulsion-based-proprietary technology to sustained deliver human bone morphogenetic protein 2 (hBMP2), a growth factor largely used for osteogenic induction. hBMP2 release profile, measured in vitro, showed a moderate "burst" release of 20% of the load in first 3 days, followed by a sustained release of 3% of the load along the following 21 days. PLGA microbeads loaded with fluorescent marker (8 mg/gPLGA ) and hydroxyapatite (30 mg/gPLGA ) were also fabricated and successfully dispersed within three-dimensional (3D) alginate scaffold (Ca-alginate 2% wt/wt) in a range between 50 and 200 mg/cm3 ; the presence of microcarriers within the scaffold induced a variation of its stiffness between 0.03 and 0.06 MPa; whereas the scaffold surface area was monitored always in the range of 190-200 m2 /g. Uniform microcarriers dispersion was obtained up to 200 mg/cm3 ; higher loading values in the 3D scaffold produced large aggregates. The release data and the surface area were, then, used to simulate by finite element modeling the hBMP2 mass transfer within the 3D hydrogel bioengineered with stem cells, in dynamic and static cultivations. The simulation was developed with COMSOL Multiphysics® giving a good representation of hBMP2 mass balances along microbeads (bulk eroded) and on cell surface (cell binding). hBMP2 degradation rate was also taken into account in the simulations. hBMP2 concentration of 20 ng/cm3 was set as a target because it has been described as the minimum effective value for stem cells stimulation versus the osteogenic phenotype. The sensitivity analysis suggested the best microbeads/cells ratio in the 3D microenvironment, along 21 days of cultivations in both static and dynamic cultivation (perfusion) conditions. The simulated formulation was so assembled experimentally using human mesenchymal stem cells and an improved scaffold stiffness up to 0.09 MPa (n = 3; p ≤ 0.01) was monitored after 21 days of cultivation; moreover a uniform extracellular matrix deposition within the 3D system was detected by Von Kossa staining, especially in dynamic conditions. The results indicated that the described tool can be useful for the design of 3D bioengineered microarchitecture by quantitative understanding.


Subject(s)
Bone Morphogenetic Protein 2 , Drug Carriers , Mesenchymal Stem Cells/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer , Stem Cell Niche/drug effects , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/pharmacokinetics , Bone Morphogenetic Protein 2/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Humans , Hydrogels/chemistry , Hydrogels/pharmacokinetics , Hydrogels/pharmacology , Mesenchymal Stem Cells/cytology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Tissue Engineering
11.
Anticancer Drugs ; 30(1): 72-80, 2019 01.
Article in English | MEDLINE | ID: mdl-30239423

ABSTRACT

Although salinomycin sodium (SS) has shown in-vitro potential to inhibit cancer stem cell growth and development, its low water solubility makes it a poor candidate as an oral chemotherapeutic agent. To improve the bioavailability of SS, SS was encapsulated here using D-α-tocopherol polyethylene glycol succinate (TPGS)-emulsified poly(lactic-co-glycolic acid) (PLGA) nanoparticles and compared with its parent SS in terms of absorption, pharmacokinetics, and efficacy in suppressing nasopharyngeal carcinomas stem cells. The pharmacokinetics of SS and salinomycin sodium-loaded D-α-tocopherol polyethylene glycol succinate-emulsified poly(lactic-co-glycolic acid) nanoparticles (SLN) prepared by nanoprecipitation were analyzed in-vivo by timed-interval blood sampling and oral administration of SS and SLN to rats. Sensitive liquid chromatography-mass spectrometry (LC-MS) was developed to quantify plasma drug concentrations. SS and SLN transport in Caco-2 cells was also investigated. The therapeutic efficacy of SS and SLN against cancer stem cells was determined by orally administering the drugs to mice bearing CNE1 and CNE2 nasopharyngeal carcinoma xenografts and then evaluating CD133 cell proportions and tumorsphere formation. The in-vivo trial with rats showed that the Cmax, AUC(0-t), and Tmax for orally administered SLN were all significantly higher than those for SS (P<0.05). These findings were corroborated by a Caco-2 cell Transwell assay showing that relative SLN absorption was greater than that of SS on the basis of their apparent permeability coefficients (Papp). Significantly, therapeutic SLN efficacy against nasopharyngeal carcinoma stem cells was superior to that of SS. TPGS-emulsified PLGA nanoparticles effectively increase SS solubility and bioavailability. SLN is, therefore, promising as an oral chemotherapeutic agent against cancer stem cells.


Subject(s)
Nanoparticles/administration & dosage , Pyrans/administration & dosage , Pyrans/pharmacokinetics , alpha-Tocopherol/administration & dosage , Animals , Caco-2 Cells , Emulsions/administration & dosage , Emulsions/pharmacokinetics , Emulsions/pharmacology , Humans , Intestinal Absorption , Male , Mice , Mice, Inbred BALB C , Nanoparticles/metabolism , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Pyrans/blood , Pyrans/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Succinates/administration & dosage , Succinates/pharmacokinetics , alpha-Tocopherol/pharmacokinetics
12.
Nanomedicine ; 21: 102073, 2019 10.
Article in English | MEDLINE | ID: mdl-31376570

ABSTRACT

Biodegradable polymeric nanoparticles (NP) made from poly (lactid-co-glycolide) acid (PLGA) and chitosan (CS) hold promise as innovative formulations for targeted delivery. Since interactions of such NP with primary human immune cells have not been characterized, yet, here we assessed the effect of PLGA or CS-PLGA NP treatment on human peripheral blood mononuclear cells (PBMC), as well as on monocyte-derived DC (moDC). Amongst PBMC, antigen presenting cells (APC) showed higher uptake of both NP preparations than lymphocytes. Furthermore, moDC internalized CS-PLGA NP more efficiently than PLGA NP, presumably because of receptor-mediated endocytosis. Consequently, CS-PLGA NP were delivered mostly to endosomal compartments, whereas PLGA NP primarily ended up in lysosomes. Thus, CS-PLGA NP confer enhanced delivery to endosomal compartments of APC, offering new therapeutic options to either induce or modulate APC function and to inhibit pathogens that preferentially infect APC.


Subject(s)
Antigen-Presenting Cells/metabolism , Chitosan , Endosomes/metabolism , Leukocytes, Mononuclear/metabolism , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Antigen-Presenting Cells/cytology , Chitosan/chemistry , Chitosan/pharmacokinetics , Chitosan/pharmacology , Humans , Leukocytes, Mononuclear/cytology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology
13.
Pharm Dev Technol ; 24(4): 395-401, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30422727

ABSTRACT

Recombinant human growth hormone (rhGH) therapy must be administered as a daily injection due to its short half-life. To achieve sustained release of rhGH, the preparation of rhGH-loaded PLGA microspheres was investigated, and the influence of various factors on encapsulation efficiency was tested, including rhGH concentration, the ratio of internal phase to organic phase, stirring speed, PVA concentration, surrounding pH value, and the type of emulsifier and organic solvent. A pharmacokinetic study was performed by subcutaneous administration to explore the sustained release effect. It was found that rhGH-loaded PLGA microspheres were prepared with a narrow size distribution, and optimization of the formulation could enhance encapsulation efficiency. FTIR analysis indicated that the activity of rhGH was maintained after encapsulation. The pharmacokinetic behavior of rhGH solutions was consistent with a two-compartment model, which showed fast absorption and distribution. RhGH-loaded PLGA microspheres achieved a higher bioavailability and a long-term effective concentration by controlling the release, and PLGA 50/50 demonstrated favorable AUC compared with PLGA 75/25. Nevertheless, the higher bioavailability of rhGH-loaded PLGA microspheres lacking Span 80 did not predicate better sustained release behavior, indicating that further investigation is needed to explore the use of bioavailability as the standard in evaluating the sustained release characteristics and in vivo behavior of microspheres.


Subject(s)
Human Growth Hormone/chemical synthesis , Human Growth Hormone/pharmacokinetics , Microspheres , Polylactic Acid-Polyglycolic Acid Copolymer/chemical synthesis , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Animals , Delayed-Action Preparations/chemical synthesis , Delayed-Action Preparations/pharmacokinetics , Humans , Male , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemical synthesis , Recombinant Proteins/pharmacokinetics
14.
AAPS PharmSciTech ; 20(8): 320, 2019 Oct 23.
Article in English | MEDLINE | ID: mdl-31646399

ABSTRACT

The in vitro drug release in an aqueous medium is a critical performance metric for a sustained release drug product. During long-term release studies, drugs may degrade in the release medium, and such degradation can lead to errors in drug release quantitation. Using dexamethasone as a model drug and LC-MS/MS methods employing dexamethasone-d4 as an internal standard, this study identified that dexamethasone can degrade into 13 major degradation products in phosphate buffered saline (PBS) as a function of time, temperature (25, 37, and 45°C), and light exposure. A putative scheme for dexamethasone degradation pathways in PBS has been proposed. In proof-of-concept studies, the analytical method was used to quantitate dexamethasone and its degradation products during in vitro release studies with sustained release dexamethasone-poly(D,L-lactide-co-glycolide) (PLGA) implants incubated in phosphate buffer saline (PBS). Further, mathematical approaches were developed to estimate drug release from implants after accounting for drug degradation in PBS. The LC-MS/MS analytical method and the mathematical approaches developed could be used for assessing the stability and/or release of dexamethasone during manufacturing, storage, and use of various dosage forms.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Dexamethasone/pharmacokinetics , Drug Delivery Systems/methods , Water/metabolism , Anti-Inflammatory Agents/administration & dosage , Chromatography, Liquid/methods , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Dexamethasone/administration & dosage , Drug Implants , Drug Liberation , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Tandem Mass Spectrometry/methods
15.
AAPS PharmSciTech ; 20(2): 64, 2019 Jan 09.
Article in English | MEDLINE | ID: mdl-30627822

ABSTRACT

Spherical poly (D, L-lactic-co-glycolic acid) microparticles (PLGA-MPs) have long been investigated in order to achieve sustained delivery of proteins/peptides. However, the formation mechanism and release characteristics of the specific shape MPs were still unknown. This study aimed to develop a novel-dimpled exenatide-loaded PLGA-MPs (Exe-PLGA-MPs) using an ultra-fine particle processing system (UPPS) and investigate the formation mechanism and release characteristics. Exe-PLGA-MPs were prepared by UPPS and optimized based on their initial burst within the first 24 h and drug release profiles. Physicochemical properties of Exe-PLGA-MPs, including morphology, particle size, and structural integrity of Exe extracted from Exe-PLGA-MPs, were evaluated. Furthermore, pharmacokinetic studies of the optimal formulation were conducted in Sprague-Dawley (SD) rats to establish in vitro-in vivo correlations (IVIVC) of drug release. Exe-PLGA-MPs with dimpled shapes and uniform particle sizes achieved a high encapsulation efficiency (EE%, 91.50 ± 2.65%) and sustained drug release for 2 months in vitro with reduced initial burst (20.42 ± 1.64%). Moreover, the pharmacokinetic studies revealed that effective drug concentration could be maintained for 3 weeks following a single injection of dimpled Exe-PLGA-MPs with high IVIVC. Dimpled PLGA-MPs prepared using the UPPS technique could thus have great potential for sustained delivery of macromolecular proteins/peptides.


Subject(s)
Chemistry, Pharmaceutical/methods , Exenatide/chemical synthesis , Microspheres , Polylactic Acid-Polyglycolic Acid Copolymer/chemical synthesis , Animals , Delayed-Action Preparations/chemical synthesis , Delayed-Action Preparations/pharmacokinetics , Drug Evaluation, Preclinical/methods , Drug Liberation , Exenatide/pharmacokinetics , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/pharmacokinetics , Male , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Rats , Rats, Sprague-Dawley
16.
Bioconjug Chem ; 29(12): 4083-4089, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30424597

ABSTRACT

Most neurodegenerative disorders are characterized by deposits of misfolded proteins and neuronal degeneration in specific brain regions. Growing evidence indicates that lysosomal impairment plays a primary pathogenic role in these diseases, in particular, the occurrence of increased lysosomal pH. Thus, therapeutic development aiming at restoring lysosomal function represents a novel, precise, and promising strategy for the treatment of these pathologies. Herein we demonstrate that acidic oil-in-water nanoemulsions loaded with poly(dl-lactide- co-glycolide) (PLGA) are able to rescue impaired lysosomal pH in genetic cellular models of Parkinson's disease. For in vivo assays, nanoemulsions were labeled with an original synthetic hydrophobic far red-emitting dye to allow fluorescence monitoring. Following stereotaxic injection in the mouse brain, widespread diffusion of the nanocarrier was observed, up to 500 µm from the injection site, as well as internalization into the lysosomal compartment in brain cells. Finally, promising preliminary assays of systemic administration demonstrate that a fraction of the formulation crosses the blood brain barrier, penetrates the brain parenchyma, is internalized by cells, and colocalizes with lysosomal markers. Overall, these results suggest the feasibility and the therapeutic potential of this new nanoformulation as an effective drug delivery tool to the brain, with the potential to rescue pathological lysosomal deficits.


Subject(s)
Hydrogen-Ion Concentration , Lysosomes/metabolism , Nanoparticles , Neurodegenerative Diseases/drug therapy , Polylactic Acid-Polyglycolic Acid Copolymer/therapeutic use , Animals , Blood-Brain Barrier , Cell Line, Tumor , Drug Carriers , Emulsions , Endocytosis , Humans , Mice , Neurodegenerative Diseases/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics
17.
Biomacromolecules ; 19(6): 2302-2307, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29742350

ABSTRACT

Polycaprolactone (PCL) was reported a long time ago; however, its biomedical applications has not been extensively investigated in comparison with poly(lactide- co-glycolide) (PLGA) due to its too slow degradation profile. Here, we are reporting an oxalate-connected oligocaprolactone multiblock copolymer (PCL-OX) as a fast degradable PCL while maintaining its crystalline properties and low melting point of PCL. The in vivo application of the paclitaxel-loaded PCL-OX microspheres provided a steady plasma drug concentration of 6-9 µg/mL over 28 days, similar to that of the PLGA microspheres. Both PCL and PLGA microspheres were completely cleared two months after in vivo implantation. The PCL-OX microspheres showed a similar tissue compatibility to that of PLGA microspheres in the subcutaneous layer of rats. These findings suggest that PCL-OX is a useful biomaterial that solves the slow degradation problems of PCL and, thus, may find uses in other biomedical applications as an alternative to PLGA.


Subject(s)
Biodegradable Plastics , Drug Delivery Systems , Microspheres , Paclitaxel , Polyesters , Animals , Biodegradable Plastics/chemical synthesis , Biodegradable Plastics/chemistry , Biodegradable Plastics/pharmacokinetics , Biodegradable Plastics/pharmacology , Drug Implants , Male , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology , Polyesters/chemistry , Polyesters/pharmacokinetics , Polyesters/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Rats , Rats, Sprague-Dawley
18.
Pharm Dev Technol ; 23(9): 857-864, 2018 Nov.
Article in English | MEDLINE | ID: mdl-27881046

ABSTRACT

Bee venom-loaded poly(lactic-co-glycolic acid) (PLGA) particles were prepared by double emulsion-solvent evaporation, and characterized for a sustained-release system. Factors such as the type of organic solvent, the amount of bee venom and PLGA, the type of PLGA, the type of polyvinyl alcohol, and the emulsification method were considered. Physicochemical properties, including the encapsulation efficiency, drug loading, particle size, zeta-potential and surface morphology were examined by Fourier transform infrared (FT-IR) spectroscopy, differential scanning calorimetry (DSC), and X-ray diffraction (XRD). The size of the bee venom-loaded PLGA particles was 500 nm (measured using sonication). Zeta-potentials of the bee venom-loaded PLGA particles were negative owing to the PLGA. FT-IR results demonstrated that the bee venom was completely encapsulated in the PLGA particles, indicated by the disappearance of the amine and amide peaks. In addition, sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis indicated that the bee venom in the bee venom-loaded PLGA particles was intact. In vitro release of the bee venom from the bee venom-loaded PLGA particles showed a sustained-release profile over 1 month. Bee venom-loaded PLGA particles can help improve patients' quality of life by reducing the number of injections required.


Subject(s)
Bee Venoms/chemical synthesis , Bee Venoms/pharmacokinetics , Drug Carriers/chemical synthesis , Drug Carriers/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/chemical synthesis , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Bee Venoms/administration & dosage , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemical synthesis , Delayed-Action Preparations/pharmacokinetics , Drug Carriers/administration & dosage , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Spectroscopy, Fourier Transform Infrared/methods , X-Ray Diffraction/methods
19.
Eur J Pharm Biopharm ; 159: 123-136, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33387633

ABSTRACT

This review discusses the physicochemical and mechanical properties of porcine gastrointestinal mucus from a rheological point of view. Considering mucus as a viscoelastic gel that functions as a biological barrier by limiting particles passage, lubricating the gastrointestinal tract, and protecting the stomach from gastric acids. The viscoelastic and protective properties of mucus are mainly produced by its mucin network, which is stabilized through electrostatic, hydrophobic and hydrogen bonding interactions. Otherwise, mucus rheology is determined by its polyanionic nature at physiological pH. At neutral pH, mucus presents a viscous behavior produced by chains crosslinking. While, at acidic pH, mucus exhibits an elastic behavior related with the extended conformation that produces mucus gelation at the stomach. Additionally, rheology studies the degree of adhesion between a polymer-mucus mixture through rheological synergism, and how it varies at different pH conditions. Finally, mucoadhesion phenomenon is exemplified with chitosan (cationic) and poly (lactic-co-glycolic) acid (anionic) polymers.


Subject(s)
Drug Carriers/chemistry , Drug Development , Gastric Mucosa/metabolism , Mucus/chemistry , Nanoparticles/chemistry , Adhesiveness , Administration, Oral , Animals , Chitosan/chemistry , Chitosan/pharmacokinetics , Drug Carriers/pharmacokinetics , Gastric Mucosa/chemistry , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Mucus/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Rheology , Swine , Viscosity
20.
Nat Nanotechnol ; 16(7): 820-829, 2021 07.
Article in English | MEDLINE | ID: mdl-33795849

ABSTRACT

The poor transport of molecular and nanoscale agents through the blood-brain barrier together with tumour heterogeneity contribute to the dismal prognosis in patients with glioblastoma multiforme. Here, a biodegradable implant (µMESH) is engineered in the form of a micrometre-sized poly(lactic-co-glycolic acid) mesh laid over a water-soluble poly(vinyl alcohol) layer. Upon poly(vinyl alcohol) dissolution, the flexible poly(lactic-co-glycolic acid) mesh conforms to the resected tumour cavity as docetaxel-loaded nanomedicines and diclofenac molecules are continuously and directly released into the adjacent tumour bed. In orthotopic brain cancer models, generated with a conventional, reference cell line and patient-derived cells, a single µMESH application, carrying 0.75 mg kg-1 of docetaxel and diclofenac, abrogates disease recurrence up to eight months after tumour resection, with no appreciable adverse effects. Without tumour resection, the µMESH increases the median overall survival (∼30 d) as compared with the one-time intracranial deposition of docetaxel-loaded nanomedicines (15 d) or 10 cycles of systemically administered temozolomide (12 d). The µMESH modular structure, for the independent coloading of different molecules and nanomedicines, together with its mechanical flexibility, can be exploited to treat a variety of cancers, realizing patient-specific dosing and interventions.


Subject(s)
Absorbable Implants , Antineoplastic Combined Chemotherapy Protocols , Brain Neoplasms/drug therapy , Polylactic Acid-Polyglycolic Acid Copolymer , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line , Diclofenac/pharmacokinetics , Diclofenac/pharmacology , Docetaxel/pharmacokinetics , Docetaxel/pharmacology , Drug Implants/pharmacokinetics , Drug Implants/pharmacology , Female , Humans , Mice , Mice, Nude , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL