Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biomacromolecules ; 17(1): 20-31, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26636723

ABSTRACT

Smart delivery system of photosensitizer chlorin e6 (Ce6) has been developed for targeted photodynamic therapy (PDT). Simple self-assemblies of the mixtures comprising soybean lecithin derived phosphatidylcholine (PC), phosphatidylethanolamine-poly(L-histidine)40 (PE-p(His)40), and folic acid (FA) conjugated phosphatidylethanolamine-poly(N-isopropylacrylamide)40 (PE-p(NIPAM)40-FA) in different ratios yield smart nanospheres characterized by (i) stable and uniform particle size (∼100 nm), (ii) positive surface charge, (iii) high hydrophobic drug (Ce6) loading efficiency up to 45%, (iv) covalently linked targeting moiety, (v) low cytotoxicity, and (vi) smartness showing p(His) block oriented pH and p(NIPAM) oriented temperature responsiveness. The Ce6-encapsulated vesicular nanospheres (Ce6@VNS) were used to confirm the efficiency of cellular uptake, intracellular distribution, and phototoxicity against KB tumor cells compared to free Ce6 at different temperature and pH conditions. The Ce6@VNS system showed significant photodynamic therapeutic efficiency on KB cells than free Ce6. A receptor-mediated inhibition study proved the site-specific delivery of Ce6 in targeted tumor cells.


Subject(s)
Nanospheres/administration & dosage , Nanospheres/chemistry , Neoplasms/drug therapy , Polymers/administration & dosage , Polymers/chemistry , Acrylamides/chemistry , Cell Line, Tumor , Chlorophyllides , Histidine/chemistry , Humans , KB Cells , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Porphyrins/chemistry
2.
J Nanosci Nanotechnol ; 14(8): 5633-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25935980

ABSTRACT

In this study, trigonelline, a niacin-related compound was incorporated into chitosan nanoparticles through ion-complex formation between anionic carboxylic acid group of trigonelline and cationic amine group of chitosan. Morphology of trigonelline-incorporated chitosan nanoparticles have spherical shape with less than 500 nm in size and thier size distribution showed quite unimodel phase. Even though trigonelline and trigonelline-incorporated chitosan nanoparticles were not significantly affected to the proliferation of tumor cells, invasion of tumor cells was effectively inhibited by trigonelline-incorporated chitosan nanoparticles. We suggested that trigonelline-incorporated chitosan nanoparticles are promising candidate for inhibition of tumor cell invasion.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Chitosan/chemistry , Nanoparticles , Alkaloids/chemistry , Cell Line, Tumor , Humans , Neoplasm Invasiveness/prevention & control
3.
Int J Nanomedicine ; 8: 809-19, 2013.
Article in English | MEDLINE | ID: mdl-23589688

ABSTRACT

PURPOSE: The aim of this study was to make 5-aminolevulinic acid (5-ALA)-incorporated nanoparticles using methoxy polyethylene glycol/chitosan (PEG-Chito) copolymer for application in photodynamic therapy for colon cancer cells. METHODS: 5-ALA-incorporated (PEG-Chito-5-ALA) nanoparticles were prepared by ion complex formation between 5-ALA and chitosan. Protoporphyrin IX accumulation in the tumor cells and phototoxicity induced by PEG-Chito-5-ALA nanoparticles were assessed using CT26 cells in vitro. RESULTS: PEG-Chito-5-ALA nanoparticles have spherical shapes with sizes diameters 200 nm. More specifically, microscopic observation revealed a core-shell structure of PEG-Chito-5-ALA nanoparticles. 1H NMR spectra showed that 5-ALA was incorporated in the core of the nanoparticles. In the absence of light irradiation, all components such as 5-ALA, empty nanoparticles, and PEG-Chito-5-ALA nanoparticles did not affect the viability of cells. However, 5-ALA or PEG-Chito-5-ALA nanoparticles induced tumor cell death under light irradiation, and the viability of tumor cells was dose-dependently decreased according to the increase in irradiation time. In particular, PEG-Chito-5-ALA nanoparticles induced increased phototoxicity and higher protoporphyrin IX accumulation into the tumor cells than did 5-ALA alone. Furthermore, PEG-Chito-5-ALA nanoparticles accelerated apoptosis/necrosis of tumor cells, compared to 5-ALA alone. CONCLUSION: PEG-Chito-5-ALA nanoparticles showed superior delivery capacity of 5-ALA and phototoxicity against tumor cells. These results show that PEG-Chito-5-ALA nanoparticles are promising candidates for photodynamic therapy of colon cancer cells.


Subject(s)
Aminolevulinic Acid/chemistry , Chitosan/analogs & derivatives , Drug Carriers/chemistry , Nanoparticles/chemistry , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Polyethylene Glycols/chemistry , Aminolevulinic Acid/administration & dosage , Aminolevulinic Acid/metabolism , Cell Line, Tumor , Chitosan/administration & dosage , Chitosan/chemistry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Carriers/administration & dosage , Humans , Particle Size , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/metabolism , Polyethylene Glycols/administration & dosage , Protoporphyrins/analysis , Protoporphyrins/metabolism
4.
Int J Nanomedicine ; 8: 485-93, 2013.
Article in English | MEDLINE | ID: mdl-23390366

ABSTRACT

PURPOSE: All-trans retinoic acid (RA)-incorporated nanoparticles were prepared using deoxycholic acid-conjugated dextran (DexDA). Anticancer activity of RA-incorporated DexDA nanoparticles were tested in vitro and in vivo. METHODS: RA-incorporated nanoparticles were prepared by dialysis. Antiproliferative and anti-invasive potential of RA-incorporated nanoparticles were studied using CT26 colorectal carcinoma cells. RESULTS: RA-incorporated nanoparticles have small particle sizes of around 70-300 nm and spherical shapes. The higher drug-feeding ratio and higher substitution degree of deoxycholic acid in the conjugates resulted in higher drug contents, lower loading efficiency, and larger particle size. RA release rate became slower at higher drug contents and higher substitution degree of deoxycholic acid in the DexDA conjugates. The antiproliferation activity, anti-invasive activity, and matrix metalloproteinase 2 expression of RA-incorporated nanoparticles against CT26 cells in vitro was similar to RA. However, RA-incorporated nanoparticles had superior antimetastatic activity in an animal pulmonary metastatic model of CT26 cells compared to RA itself. CONCLUSION: RA-incorporated nanoparticles showed similar anticancer activity in vitro and superior antimetastatic activity in vivo in a pulmonary metastatic model of CT26 cells. We suggest that RA-incorporated nanoparticles are promising vehicles for efficient delivery of RA.


Subject(s)
Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Deoxycholic Acid/administration & dosage , Dextrans/chemistry , Nanoparticles/administration & dosage , Tretinoin/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Deoxycholic Acid/chemistry , Mice , Nanoparticles/chemistry , Neoplasm Invasiveness , Neoplasm Metastasis , Particle Size , Polymers/administration & dosage , Polymers/chemistry , Polymers/pharmacokinetics , Tretinoin/chemistry , Tretinoin/pharmacokinetics
5.
Eur J Pharm Biopharm ; 85(3 Pt A): 503-10, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23429232

ABSTRACT

Hexyl-aminolevulinic acid (HALA) was compared with aminolevulinic acid (ALA) in terms of improving ALA-based photodynamic therapy (PDT) for human intra- and extrahepatic cholangiocarcinoma (CCA) HuCC-T1 and SNU1196 cells. Because of the different uptake mechanisms of HALA, a relatively higher amount of protoporphyrin IX (PpIX) was induced in the both CCA cell types at low concentrations of HALA. Furthermore, higher expression of porphobilinogen deaminase, coproporphyrinogen III oxidase, and protoporphyrinogen oxidase, the key enzymes for synthesizing PpIX in the heme biosynthetic pathway, facilitated the exuberant generation of PpIX in HuCC-T1 cells. PpIX accumulation with ALA was markedly different between the two CCA cell types. Even at lower concentrations of ALA, SNU1196 cell successfully synthesized PpIX, due to the higher expression of the ALA transporter, mammalian H (+)/peptide co-transporter PEPT1. Considering the difference of PEPT1 or key enzyme expression, HALA could be a very effective substitute for ALA in doing PDT for cure of CCA.


Subject(s)
Aminolevulinic Acid/analogs & derivatives , Cholangiocarcinoma/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Aminolevulinic Acid/administration & dosage , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/pharmacology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cell Line, Tumor , Cholangiocarcinoma/pathology , Dose-Response Relationship, Drug , Humans , Peptide Transporter 1 , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Protoporphyrins/metabolism , Symporters/genetics
6.
Int J Pharm ; 422(1-2): 454-61, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22093956

ABSTRACT

The aim of this study is to investigate antitumor activity of all-trans retinoic acid (RA)-incorporated glycol chitosan (GC) nanoparticles. RA-incorporated GC nanoparticles were prepared by electrostatic interaction between RA and amine group of GC. RA-incorporated GC nanoparticles have spherical shape and their particle size was 317 ± 34.5 nm. They were simply reconstituted into aqueous solution without changes of intrinsic properties. RA-incorporated GC nanoparticles were evidently inhibited the proliferation of HuCC-T1 cholangiocarcinoma cells at higher than 20 µg/ml of RA concentration while empty GC vegicles did not affect to the viablity of tumor cells. Apoptosis and necrosis analysis of tumor cells with treatment of RA or RA-incorporated GC nanoparticles also supported these results. Invasion test using Matrigel also showed that invasion of tumor cells was significantly inhibited at higher than 20 µg/ml of RA concentration. Wound healing assay also showed that RA-incorporated GC nanoparticles were inhibited migration of tumor cells as similar to RA itself. Our results suggested that RA-incorporated GC nanoparticles is a promising vehicles for RA delivery to HuCC-T1 cholangiocarcinoma cells.


Subject(s)
Antineoplastic Agents/pharmacology , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/drug effects , Chitosan/chemistry , Cholangiocarcinoma/pathology , Drug Carriers , Nanoparticles , Nanotechnology , Technology, Pharmaceutical/methods , Tretinoin/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Bile Ducts, Intrahepatic/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Drug Compounding , Humans , Necrosis , Neoplasm Invasiveness , Particle Size , Tretinoin/chemistry
7.
Eur J Pharm Biopharm ; 80(2): 453-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22024407

ABSTRACT

Photodynamic therapy (PDT) is a palliative therapy and has been used to cure cholangiocarcinoma (CC), which has a poor prognosis and limited available curative therapy. PDT was shown to improve the median survival time of advanced-stage patients. Recently, 5-aminolevulinic acid (ALA) has been used as a pro-photosensitizer, which can be transferred to intercellular protoporphyrin IX (PpIX), which is a strong photosensitizer, via the heme pathway. The main limitation of using ALA in PDT is the hydrophilic properties of ALA, which results in low cellular uptake. In this study, non-ionic surfactants, pluronic F68 (PF68) and Tween 80 (TW80), were used to address this limitation. The human CC cell line, HuCC-T1, was cotreated with ALA and different concentrations of surfactants for 4h. The effect of surfactants was evaluated by monitoring the uptake of ALA, the fluorescence intensity of PpIX, and the cell survival rate after suitable light irradiation. Cotreatment with the surfactant resulted in an increased intracellular ALA level, PpIX formation, and phototoxicity.


Subject(s)
Aminolevulinic Acid/pharmacology , Cholangiocarcinoma/drug therapy , Photosensitizing Agents/pharmacology , Surface-Active Agents/chemistry , Aminolevulinic Acid/administration & dosage , Aminolevulinic Acid/pharmacokinetics , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/drug effects , Bile Ducts, Intrahepatic/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cholangiocarcinoma/pathology , Excipients/chemistry , Humans , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics , Poloxamer/chemistry , Polysorbates/chemistry , Protoporphyrins/metabolism
8.
Colloids Surf B Biointerfaces ; 88(2): 711-6, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21872455

ABSTRACT

In this study, porous sodium hyaluronic acid/sodium alginate (HA/SA) scaffold based on interpenetrating polymeric network (IPN) technique has been fabricated, where HA and SA were cross-linked with poly(ethylene glycol) diglycidyl ether (PEGDG) and calcium chloride, respectively. The mean pore size and the swelling ratio of fabricated scaffolds decreased, and the compressive strength increased as the content of SA increased in HA/SA IPN scaffold. Rabbit chondrocytes were seeded within the HA/SA IPN scaffolds, and then their proliferation as well as chondrogenic differentiation was examined. DNA contents observed from the chondrocytes cultured in the IPN scaffolds increased with time over 21 days, which demonstrated that the rabbit chondrocytes continued to proliferate in HA/SA scaffolds. Results of the 1,9-dimethylmethylene blue (DMMB) and p-dimethylaminobenzaldehyde (DMBA) assays showed that glycosaminoglycan (s-GAG) and collagen contents increased over culture period, indicating the chondrogenic differentiation in the scaffold. Reverse transcription-polymerase chain reaction (RT-PCR) results showed the expression of type II collagen, the main chondrogenic differentiation marker. The bands indicating mRNA expression of type II collagen increased with the culture period. These results demonstrated that the porous HA/SA IPN scaffolds were successfully prepared and could serve as an effective delivery system of the three-dimensional culture of chondrocytes.


Subject(s)
Alginates/chemistry , Chondrocytes/cytology , Hyaluronic Acid/chemistry , Tissue Scaffolds/chemistry , Animals , Calcium Chloride/chemistry , Cell Differentiation , Cells, Cultured , Chondrocytes/metabolism , Collagen/metabolism , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Methylene Blue/analogs & derivatives , Methylene Blue/chemistry , Rabbits , Reverse Transcriptase Polymerase Chain Reaction
9.
Arch Pharm Res ; 34(1): 159-67, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21468928

ABSTRACT

In this study, we synthesized deoxycholic acid (DA)-conjugated dextran (DexDA) and prepared doxorubicin (DOX)-encapsulated nanoparticles using DexDA conjugates. Since DexDA conjugates have amphiphilic properties, they will show self-aggregation behavior at aqueous environment. To approve self-aggregation behavior, critical aggregation concentration value of DexDA conjugates was evaluated using fluorescence spectroscopy. DOX-incorporated DexDA nanoparticles were less than 200 nm. The higher substitution degree of DA and higher drug feeding ratio resulted in increased particle size. Drug release was decreased by increase of substitution degree value of DA and increase of drug feeding ratio. At in vitro cytotoxicity test using DOX-resistant CT26 colon carcinoma cells, higher antitumor activity was obtained with DOX-incorporated nanoparticles compared to free DOX. Fluorescence microscopic observation verified this result, i.e. nanoparticles were properly entered into tumors cells and maintained longer compared to DOX itself. These results suggested that DOX-incorporated DexDA nanoparticles are promising vehicles for antitumor drug delivery.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Colonic Neoplasms/drug therapy , Deoxycholic Acid/chemistry , Doxorubicin/pharmacology , Animals , Antibiotics, Antineoplastic/administration & dosage , Cell Line, Tumor , Colonic Neoplasms/pathology , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Mice , Nanoparticles , Particle Size , Spectrometry, Fluorescence
10.
Korean J Anesthesiol ; 57(3): 364-366, 2009 Sep.
Article in English | MEDLINE | ID: mdl-30625888

ABSTRACT

Cauda equina syndrome is a well-known but rare complication of spinal anesthesia. An 80-year-old man was scheduled for both herniorrhaphy. Spinal anesthesia was performed at the L3-4 interspinous space with 0.5% hyperbaric bupivacaine 12 mg. Eight hours after anesthesia, the patient complained bilateral sensorimotor deficits of the lower extremities and peroneal region. Urinary and fecal incontinence were also observed. MRI and myelography showed severe central spinal stenosis at L3-4 and L4-5. EMG showed cauda equina syndrome. Seven weeks after the procedure, left decompressive subtotal laminectomy L2-L5 was done. The patient still complains the neuropathic pain in the both lower extremities and ambulates using a walker. The local anesthetic was injected into thecal sac between maximum stenoses, and it is likely that there was poor upward spread leading to maldistribution of local anesthetic and resultant local anesthetic toxicity.

11.
Korean J Anesthesiol ; 57(4): 444-449, 2009 Oct.
Article in English | MEDLINE | ID: mdl-30625904

ABSTRACT

BACKGROUND: A priming dose of rocuronium can shorten the onset time of neuromuscular blockade. The purpose of this study was to evaluate the effect of priming with rocuronium on the onset time and intubation conditions during tracheal intubation with low-dose rocuronium (0.35 mg/kg) and to compare results with those for rocuronium 0.45 mg/kg. METHODS: One hundred twenty four patients were randomly allocated to three groups. Following induction of anesthesia, groups I and III received normal saline while group II received a priming dose of rocuronium (0.05 mg/kg). Three minutes after priming, groups I, II and III received, respectively, 0.45 mg/kg, 0.3 mg/kg and 0.35 mg/kg rocuronium. Intubation was performed 2 minutes after the administration of an intubating dose and intubation conditions were evaluated. Neuromuscular blockade was assessed by accelerography. RESULTS: The proportion of cases having optimal intubation conditions in group I was higher than in groups II and III. There was no significant difference in the onset times among groups. Neuromuscular blockade at 60, 90 and 120 seconds after an intubating dose was similar among all groups except at 60 sec. Maximal blockade for group I was deep compared to groups II and III. CONCLUSIONS: Rocuronium 0.35 mg/kg does not provide satisfactory intubation conditions. There are no effects on onset time and intubation conditions due to priming during tracheal intubation with rocuronium 0.35 mg/kg.

SELECTION OF CITATIONS
SEARCH DETAIL
...