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










Publication year range
1.
Biomaterials ; 35(2): 836-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24144908

ABSTRACT

Nanoparticle-mediated delivery of chemotherapies has demonstrated enhanced anti-cancer efficacy, mainly through the mechanisms of both passive and active targeting. Herein, we report other than these well-elucidated mechanisms, rationally designed nanoparticles can efficiently deliver drugs to cancer stem cells (CSCs), which in turn contributes significantly to the improved anti-cancer efficacy. We demonstrate that doxorubicin-tethered gold nanoparticles via a poly(ethylene glycol) spacer and an acid-labile hydrazone bond mediate potent doxorubicin delivery to breast CSCs, which reduces their mammosphere formation capacity and their cancer initiation activity, eliciting marked enhancement in tumor growth inhibition in murine models. The drug delivery mediated by the nanoparticles also markedly attenuates tumor growth during off-therapy stage by reducing breast CSCs in tumors, while the therapy with doxorubicin alone conversely evokes an enrichment of breast CSCs. Our findings suggest that with well-designed drug delivery system, the conventional chemotherapeutic agents are promising for cancer stem cell therapy.


Subject(s)
Doxorubicin/pharmacology , Drug Delivery Systems , Gold/chemistry , Nanoparticles/chemistry , Neoplastic Stem Cells/drug effects , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred NOD , Polyethylene Glycols/chemistry
2.
Biomater Sci ; 1(11): 1143-1150, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-32481937

ABSTRACT

Effective systemic therapy is often necessary to treat hepatocellular carcinoma (HCC). We synthesized a Gal-PPE nanogel consisting of a cross-linked polyphosphate core and galactosylated poly(ethylene glycol) arms for enhanced doxorubicin delivery to diethylnitrosamine-induced HCC in rats. The Gal-PPE nanogel exhibited high affinity to HepG2 cells in vitro, mediated by the asialoglycoprotein receptor. In vivo studies revealed that the Gal-PPE nanogel was taken up more efficiently by hepatocytes, in contrast to m-PPE nanogel. Consequently, doxorubicin delivery with Gal-PPE significantly inhibited the progress of HCC, reducing neoplastic liver nodules and prolonging the survival time of HCC rats more significantly. These results demonstrate the potential of Gal-PPE as a nanocarrier for improved HCC chemotherapy.

3.
J Control Release ; 161(3): 875-83, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22762887

ABSTRACT

The targeted delivery of small interfering RNA (siRNA) to specific tumor tissues and tumor cells remains as one of the key challenges in the development of RNA interference as a therapeutic application. To target breast cancer, we developed a therapeutic delivery system using a fusion protein of an anti-Her2 single-chain antibody fragment with a positively charged protamine, namely F5-P, as the carrier to specifically deliver siRNA-targeting DNA methyltransferases 1 and/or 3b genes (siDNMTs) into Her2-expressing breast tumor cells. The carrier F5-P, expressed by the Escherichia coli system, was able to bind siRNA molecules and specifically deliver the siRNA to Her2-expressing BT474 breast cancer cells but not Her2-nonexpressing MDA-MB-231 breast cancer cells, while delivery of siDNMTs to BT474 cells successfully silenced the expression of targeted DNA methyltransferases (DNMTs) and facilitated the de-methylation of the RASSF1A tumor suppressor gene promoter, leading to the suppression of tumor cell proliferation. Moreover, as demonstrated in the BT474 xenograft murine model, F5-P successfully delivered siRNA into a Her2-expressing breast tumor, and tumor growth inhibition was mediated by an intravenous injection of F5-P/siDNMTs complex by down-regulating the expression of DNMTs and restoring tumor suppressor gene expression. These data suggest that the delivery of siDNMTs by F5-P could be used to treat Her2-expressing breast cancer.


Subject(s)
Breast Neoplasms/therapy , DNA Modification Methylases/genetics , Immunoglobulin Fragments/administration & dosage , Protamines/administration & dosage , RNA, Small Interfering/administration & dosage , Receptor, ErbB-2/immunology , Animals , Cell Line, Tumor , DNA Modification Methylases/metabolism , Female , Gene Silencing , Humans , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/administration & dosage , Xenograft Model Antitumor Assays
4.
Sci Transl Med ; 4(130): 130ra48, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22517885

ABSTRACT

A major obstacle to developing small interfering RNAs (siRNAs) as cancer drugs is their intracellular delivery to disseminated cancer cells. Fusion proteins of single-chain fragmented antibodies (ScFvs) and positively charged peptides deliver siRNAs into specific target cells. However, the therapeutic potential of ScFv-mediated siRNA delivery has not been evaluated in cancer. Here, we tested whether Polo-like kinase 1 (PLK1) siRNAs complexed with a Her2-ScFv-protamine peptide fusion protein (F5-P) could suppress Her2(+) breast cancer cell lines and primary human cancers in orthotopic breast cancer models. PLK1-siRNAs transferred by F5-P inhibited target gene expression, reduced proliferation, and induced apoptosis of Her2(+) breast cancer cell lines and primary human cancer cells in vitro without triggering an interferon response. Intravenously injected F5-P/PLK1-siRNA complexes concentrated in orthotopic Her2(+) breast cancer xenografts and persisted for at least 72 hours, leading to suppressed PLK1 gene expression and tumor cell apoptosis. The intravenously injected siRNA complexes retarded Her2(+) breast tumor growth, reduced metastasis, and prolonged survival without evident toxicity. F5-P-mediated delivery of a cocktail of PLK1, CCND1, and AKT siRNAs was more effective than an equivalent dose of PLK1-siRNAs alone. These data suggest that F5-P could be used to deliver siRNAs to treat Her2(+) breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Cycle Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Receptor, ErbB-2/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
5.
J Control Release ; 156(2): 203-11, 2011 Dec 10.
Article in English | MEDLINE | ID: mdl-21839126

ABSTRACT

Delivery of small interfering RNA (siRNA) has been one of the major hurdles for the application of RNA interference in therapeutics. Here, we describe a cationic lipid assisted polymeric nanoparticle system with stealthy property for efficient siRNA encapsulation and delivery, which was fabricated with poly(ethylene glycol)-b-poly(d,l-lactide), siRNA and a cationic lipid, using a double emulsion-solvent evaporation technique. By incorporation of the cationic lipid, the encapsulation efficiency of siRNA into the nanoparticles could be above 90% and the siRNA loading weight ratio was up to 4.47%, while the diameter of the nanoparticles was around 170 to 200nm. The siRNA retained its integrity within the nanoparticles, which were effectively internalized by cancer cells and escaped from the endosome, resulting in significant gene knockdown. This effect was demonstrated by significant down-regulation of luciferase expression in HepG2-luciferase cells which stably express luciferase, and suppression of polo-like kinase 1 (Plk1) expression in HepG2 cells, following delivery of specific siRNAs by the nanoparticles. Furthermore, the nanoparticles carrying siRNA targeting the Plk1 gene were found to induce remarkable apoptosis in both HepG2 and MDA-MB-435s cancer cells. Systemic delivery of specific siRNA by nanoparticles significantly inhibited luciferase expression in an orthotopic murine liver cancer model and suppressed tumor growth in a MDA-MB-435s murine xenograft model, suggesting its therapeutic promise in disease treatment.


Subject(s)
Nanoparticles/chemistry , Neoplasms/therapy , Polyethylene Glycols/chemistry , RNA Interference , RNA, Small Interfering/administration & dosage , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Genetic Therapy , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/ultrastructure , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Polo-Like Kinase 1
6.
Bioconjug Chem ; 22(10): 1939-45, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-21866903

ABSTRACT

Multidrug resistance (MDR) is a major impediment to the success of cancer chemotherapy. The intracellular accumulation of drug and the intracellular release of drug molecules from the carrier could be the most important barriers for nanoscale carriers in overcoming MDR. We demonstrated that the redox-responsive micellar nanodrug carrier assembled from the single disulfide bond-bridged block polymer of poly(ε-caprolactone) and poly(ethyl ethylene phosphate) (PCL-SS-PEEP) achieved more drug accumulation and retention in MDR cancer cells. Such drug carrier rapidly released the incorporated doxorubicin (DOX) in response to the intracellular reductive environment. It therefore significantly enhanced the cytotoxicity of DOX to MDR cancer cells. It was demonstrated that nanoparticular drug carrier with either poly(ethylene glycol) or poly(ethyl ethylene phosphate) (PEEP) shell increased the influx but decreased the efflux of DOX by the multidrug resistant MCF-7/ADR breast cancer cells, in comparison with the direct incubation of MCF-7/ADR cells with DOX, which led to high cellular retention of DOX. Nevertheless, nanoparticles bearing PEEP shell exhibited higher affinity to the cancer cells. The shell detachment of the PCL-SS-PEEP nanoparticles caused by the reduction of intracellular glutathione significantly accelerated the drug release in MCF-7/ADR cells, demonstrated by the flow cytometric analyses, which was beneficial to the entry of DOX into the nuclei of MCF-7/ADR cells. It therefore enhanced the efficiency in overcoming MDR of cancer cells, which renders the redox-responsive nanoparticles promising in cancer therapy.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Disulfides/chemistry , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Nanoparticles/chemistry , Polyesters/chemistry , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Female , Humans , Oxidation-Reduction
7.
ACS Nano ; 5(5): 3679-92, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21462992

ABSTRACT

Multidrug resistance (MDR) is a major impediment to the success of cancer chemotherapy. Through the development of a drug delivery system that tethers doxorubicin onto the surface of gold nanoparticles with a poly(ethylene glycol) spacer via an acid-labile linkage (DOX-Hyd@AuNPs), we have demonstrated that multidrug resistance in cancer cells can be significantly overcome by a combination of highly efficient cellular entry and a responsive intracellular release of doxorubicin from the gold nanoparticles in acidic organelles. DOX-Hyd@AuNPs achieved enhanced drug accumulation and retention in multidrug resistant MCF-7/ADR cancer cells when it was compared with free doxorubicin. It released doxorubicin in response to the pH of acidic organelles following endocytosis, opposite to the noneffective drug release from doxorubicin-tethered gold nanoparticles via the carbamate linkage (DOX-Cbm@AuNPs), which was shown by the recovered fluorescence of doxorubicin from quenching due to the nanosurface energy transfer between the doxorubicinyl groups and the gold nanoparticles. DOX-Hyd@AuNPs therefore significantly enhanced the cytotoxicity of doxorubicin and induced elevated apoptosis of MCF-7/ADR cancer cells. With a combined therapeutic potential and ability to probe drug release, DOX-Hyd@AuNPs represent a model with dual roles in overcoming MDR in cancer cells and probing the intracellular release of drug from its delivery system.


Subject(s)
Doxorubicin/administration & dosage , Drug Resistance, Multiple , Gold/chemistry , Nanocapsules/chemistry , Neoplasms, Experimental/drug therapy , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Doxorubicin/chemistry , Humans , Nanocapsules/ultrastructure , Neoplasms, Experimental/pathology
8.
Biomaterials ; 32(11): 3124-33, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21277018

ABSTRACT

One of the key challenges in the development of RNA interference-based cancer therapy is the lack of an efficient delivery system for synthetic small interfering RNAs (siRNAs) that would enable efficient uptake by tumor cells and allow for significant knockdown of a target transcript in vivo. Here, we describe a micelleplex system based on an amphiphilic and cationic triblock copolymer, which can systemically deliver siRNA targeting the acid ceramidase (AC) gene for cancer therapy. This triblock copolymer, consisting of monomethoxy poly(ethylene glycol), poly(ε-caprolactone) and poly(2-aminoethyl ethylene phosphate), self-assembles into micellar nanoparticles (MNPs) in aqueous solution with an average diameter of 60 nm and a zeta potential of approximately 48 mV. The resulting micelleplex, formed by the interaction of MNPs and siRNA, was effectively internalized by BT474 breast cancer cells and siRNA was subsequently released, resulting in significant gene knockdown. This effect was demonstrated by significant down-regulation of luciferase expression in BT474-luciferase cells which stably express luciferase, and suppression of AC expression in BT474 cells at both the transcriptional and protein level, following delivery of specific siRNAs by the micelleplex. Furthermore, a micelleplex carrying siRNA targeting the AC (micelleplex(siAC)) gene was found to induce remarkable apoptosis and reduce the proliferation of cancer cells. Systemic delivery of micelleplex(siAC) significantly inhibited tumor growth in a BT474 xenograft murine model, with depressed expression of AC and no positive activation of the innate immune response, suggesting therapeutic promise for micelleplex siRNA delivery in cancer therapy.


Subject(s)
Biocompatible Materials/chemistry , Breast Neoplasms/therapy , Nanoparticles/chemistry , Polymers/chemistry , RNA, Small Interfering/genetics , Acid Ceramidase/genetics , Animals , Apoptosis/genetics , Blotting, Western , Breast Neoplasms/enzymology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Micelles , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Reverse Transcriptase Polymerase Chain Reaction
9.
ACS Nano ; 5(2): 1483-94, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21204585

ABSTRACT

Combination of two or more therapeutic strategies with different mechanisms can cooperatively prohibit cancer development. Combination of chemotherapy and small interfering RNA (siRNA)-based therapy represents an example of this approach. Hypothesizing that the chemotherapeutic drug and the siRNA should be simultaneously delivered to the same tumoral cell to exert their synergistic effect, the development of delivery systems that can efficiently encapsulate two drugs and successfully deliver payloads to targeted sites via systemic administration has proven to be challenging. Here, we demonstrate an innovative "two-in-one" micelleplex approach based on micellar nanoparticles of a biodegradable triblock copolymer poly(ethylene glycol)-b-poly(ε-caprolactone)-b-poly(2-aminoethyl ethylene phosphate) to systemically deliver the siRNA and chemotherapeutic drug. We show clear evidence that the micelleplex is capable of delivering siRNA and paclitaxel simultaneously to the same tumoral cells both in vitro and in vivo. We further demonstrate that systemic administration of the micelleplex carrying polo-like kinase 1 (Plk1) specific siRNA and paclitaxel can induce a synergistic tumor suppression effect in the MDA-MB-435s xenograft murine model, requiring a thousand-fold less paclitaxel than needed for paclitaxel monotherapy delivered by the micelleplex and without activation of the innate immune response or generation of carrier-associated toxicity.


Subject(s)
Micelles , Neoplasms/genetics , Neoplasms/metabolism , Paclitaxel/metabolism , Paclitaxel/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Animals , Base Sequence , Cell Cycle Proteins/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Synergism , Female , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Nanoparticles/chemistry , Neoplasms/pathology , Polymers/chemistry , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Time Factors , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
10.
Mol Pharm ; 8(1): 250-9, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21138272

ABSTRACT

MiRNAs are viable therapeutic targets for cancer therapy, but the targeted delivery of miRNA or its anti-miRNA antisense oligonucleotides (AMOs) remains a challenge. We report here a PEGylated LPH (liposome-polycation-hyaluronic acid) nanoparticle formulation modified with cyclic RGD peptide (cRGD) for specific and efficient delivery of AMO into endothelial cells, targeting α(v)ß3 integrin present on the tumor neovasculature. The nanoparticles effectively delivered anti-miR-296 AMO to the cytoplasm and downregulated the target miRNA in human umbilical vein endothelial cells (HUVECs), which further efficiently suppressed blood tube formulation and endothelial cell migration, owing to significant upregulation of hepatocyte growth factor-regulated tyrosine kinase substrate (HGS), whereas nanoparticles without cRGD modification showed only little AMO uptake and miRNA silencing activity. In vivo assessment of angiogenesis using Matrigel plug assay also demonstrated that cRGD modified LPH nanoparticles have potential for antiangiogenesis in miRNA therapeutics. With the delivery of anti-miR-296 AMO by targeted nanoparticles, significant decrease in microvessel formulation within Matrigel was achieved through suppressing the invasion of CD31-positive cells into Matrigel and prompting HGS expression in angiogenic endothelial cells.


Subject(s)
MicroRNAs/genetics , Nanoparticles/chemistry , Oligonucleotides, Antisense/genetics , Oligopeptides/chemistry , Animals , Blotting, Western , Cell Line , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Female , Humans , Liposomes/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Theoretical , Phosphoproteins/genetics , Phosphoproteins/metabolism
12.
Small ; 6(2): 239-46, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19924738

ABSTRACT

An efficient and safe delivery system for small interfering RNA (siRNA) is required for clinical application of RNA interfering therapeutics. Polyethyleneimine (PEI)-capped gold nanoparticles (AuNPs) are successfully manufactured using PEI as the reductant and stabilizer, which bind siRNA at an appropriate weight ratio by electrostatic interaction and result in well-dispersed nanoparticles with uniform structure and narrow size distribution. With siRNA binding, PEI-capped AuNPs induce more significant and enhanced reduction in targeted green fluorescent protein expression in MDA-MB-435s cells, though more internalized PEI/siRNA complexes in cells are evidenced by confocal laser scanning microscopy observation and fluorescence-activated cell sorting analyses. PEI-capped AuNPs/siRNA targeting endogenous cell-cycle kinase, an oncogene polo-like kinase 1 (PLK1), display significant gene expression knockdown and induce enhanced cell apoptosis, whereas it is not obvious when the cells are treated with PLK1 siRNA using PEI as the carrier. Without exhibiting cellular toxicity, PEI-capped AuNPs appear to be suitable as a potential carrier for intracellular siRNA delivery.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Polyethyleneimine/chemistry , RNA, Small Interfering/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Gene Silencing/drug effects , Gene Transfer Techniques , Gold/toxicity , Green Fluorescent Proteins/metabolism , Humans , Metal Nanoparticles/toxicity , Polyethyleneimine/toxicity , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Polo-Like Kinase 1
13.
Macromol Rapid Commun ; 31(13): 1201-6, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-21590876

ABSTRACT

Reversibly cross-linked core-shell-corona micelles based on a triblock copolymer composed of poly(aliphatic ester), polyphosphoester, and poly(ethylene glycol) are reported. The triblock copolymer is synthesized through consecutive ring-opening polymerization of ε-caprolactone and 2,4-dinitrophenylthioethyl ethylene phosphate, followed by conjugation of poly(ethylene glycol). After deprotection under mild conditions, the amphiphilic polymer forms core-shell-corona micelles with free thiols in the shell. Cross-linking of the micelles within the shell reduces their critical micellization concentration and enhances their stability against severe conditions. The redox-sensitive cross-linkage allows the facilitated release of entrapped anticancer drugs in the cytoplasm in response to the intracellular reductive environment. With enhanced stability during circulation after administration, and accelerated intracellular drug release at the target site, the biocompatible and biodegradable shell-cross-linked polymeric micelle is promising as a drug vehicle for cancer chemotherapy.

14.
Biomacromolecules ; 10(8): 2213-20, 2009 Aug 10.
Article in English | MEDLINE | ID: mdl-19586040

ABSTRACT

Surface modification is often needed in tissue engineering to enhance the interaction between cells and synthetic materials and improve the cytocompatibility and cellular functions. In this study, block copolymers of poly(L-lactic acid) and poly(ethyl ethylene phosphate) (PLLA-b-PEEP) were synthesized and used to modify the PLLA surface via a spin-coating process, to understand whether surface modification with polyphosphoester-based polymer will be osteoinductive for potential bone tissue engineering applications. X-ray photoelectron spectra measurements revealed that phosphorus atomic compositions after surface modification increased from 2.09% to 4.39% with increasing PEEP length of PLLA-b-PEEP from 58 to 224 units, which also led to a more hydrophilic surface property compared with unmodified PLLA. The initial osteoblast attachment and proliferation on the modified surfaces were significantly enhanced. Moreover, cellular alkaline phosphatase activity and mineral calcium depositions were also promoted by PEEP modification. The gene expression determined by reverse transcription polymerase chain reaction further revealed that type I collagen and osteocalcin expression were upregulated in osteoblasts cultured on the modified surfaces, indicating that PEEP modification might be potentially osteoinductive and favorable for further application in bone tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Cell Proliferation , Esters/chemistry , Osteoblasts/cytology , Osteoblasts/metabolism , Polymers/chemistry , Alkaline Phosphatase/metabolism , Animals , Animals, Newborn , Biocompatible Materials/chemical synthesis , Calcium/metabolism , Cell Adhesion , Collagen Type I/genetics , Collagen Type I/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , Polymers/chemical synthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tissue Engineering
15.
Biomaterials ; 29(32): 4348-55, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18715636

ABSTRACT

A novel amphiphilic and cationic triblock copolymer consisting of monomethoxy poly(ethylene glycol), poly(epsilon-caprolactone) (PCL) and poly(2-aminoethyl ethylene phosphate) denoted as mPEG(45)-b-PCL(100)-b-PPEEA(12) was designed and synthesized for siRNA delivery. The copolymers were well characterized by (1)H NMR spectroscopy and gel permeation chromatography. Micelle nanoparticles' (MNPs) formation of this amphiphilic copolymer in aqueous solution was studied by dynamic light scattering, transmission electron microscopy and fluorescence technique. MNPs took uniform spherical morphology with zeta potential of around 45 mV and were stabilized by hydrophobic-hydrophobic interaction in the PCL core, exhibiting the critical micelle concentration at 2.7 x 10(-3) mg/mL. Such MNPs allowed siRNA loading post nanoparticle formation without change in uniformity. The average diameter of nanoparticles after siRNA binding ranged from 98 to 125 nm depending on N/P ratios. The siRNA loaded nanoparticles can be effectively internalized and subsequently release siRNA in HEK293 cells, resulting in significant gene knockdown activities, which was demonstrated by delivering two siRNAs targeting green fluorescence protein (GFP). It effectively silenced GFP expression in 40-70% GFP-expressed HEK293 cells and it was observed that higher N/P ratio resulted in more effective silence which was likely due to better cell internalization at higher N/P ratio. MTT assay demonstrated that neither MNPs themselves nor siRNA loaded MNPs showed cytotoxicity even at high concentrations. Such cationic MNPs made from biocompatible and biodegradable polymers are promising for siRNA delivery.


Subject(s)
Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Drug Carriers/chemistry , Hydrophobic and Hydrophilic Interactions , Micelles , Polymers/chemistry , RNA, Small Interfering/chemistry , Cations/chemistry , Cell Line , Drug Delivery Systems , Gene Expression , Humans , Magnetic Resonance Spectroscopy , Microscopy, Electron, Transmission , Molecular Structure , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Water/chemistry
16.
J Control Release ; 128(1): 32-40, 2008 May 22.
Article in English | MEDLINE | ID: mdl-18395283

ABSTRACT

Cellular specific micellar systems from functional amphiphilic block copolymers are attractive for targeted intracellular drug delivery. In this study, we developed reactive micelles based on diblock copolymer of poly(ethyl ethylene phosphate) and poly(epsilon-caprolactone). The micelles were further surface conjugated with galactosamine to target asialoglycoprotein receptor (ASGP-R) of HepG2 cells. The size of micellar nanoparticles was about 70nm in diameter, and nanoparticles were negatively charged in aqueous solution. Through recognition between galactose ligands with ASGP-R of HepG2 cells, cell surface binding and internalization of galactosamine-conjugated micelles were significantly promoted, which were demonstrated by flow cytometric analyses using rhodamine 123 fluorescent dye. Paclitaxel-loaded micelles with galactose ligands exhibited comparable activity to free paclitaxel in inhibiting HepG2 cell proliferation, in contrast to the poor inhibition activity of micelles without galactose ligands particularly at lower paclitaxel doses. In addition, population of HepG2 cells arrested in G2/M phase was in positive response to paclitaxel dose when cells were incubated with paclitaxel-loaded micelles with galactosamine conjugation, which was against the performance of micelles without galactose ligand, owing to the ligand-receptor interaction. The surface functionalized micellar system is promising for specific anticancer drug transportation and intracellular drug release.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Asialoglycoprotein Receptor/metabolism , Drug Carriers/pharmacology , Paclitaxel/pharmacology , Polyesters/pharmacology , Polyethylenes/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Carriers/metabolism , Galactosamine/chemistry , Glucosamine/chemistry , Humans , Micelles , Paclitaxel/chemistry , Polyesters/chemistry , Polyesters/metabolism , Polyethylenes/chemistry , Polyethylenes/metabolism
17.
Biomacromolecules ; 9(1): 388-95, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18081252

ABSTRACT

A series of novel amphiphilic triblock copolymers of poly(ethyl ethylene phosphate) and poly(-caprolactone) (PEEP-PCL-PEEP) with various PEEP and PCL block lengths were synthesized and characterized. These triblock copolymers formed micelles composed of a hydrophobic core of poly(-caprolactone) (PCL) and a hydrophilic shell of poly(ethyl ethylene phosphate) (PEEP) in aqueous solution. The micelle morphology was spherical, determined by transmission electron microscopy. It was found that the size and critical micelle concentration values of the micelles depended on both hydrophobic PCL block length and PEEP hydrophilic block length. The in vitro degradation characteristics of the triblock copolymers were investigated in micellar form, showing that these copolymers were completely biodegradable under enzymatic catalysis of Pseudomonas lipase and phosphodiesterase I. These triblock copolymers were used for paclitaxel (PTX) encapsulation to demonstrate the potential in drug delivery. PTX was successfully loaded into the micelles, and the in vitro release profile was found to be correlative to the polymer composition. These biodegradable triblock copolymer micelles are potential as novel carriers for hydrophobic drug delivery.


Subject(s)
Drug Carriers , Micelles , Polymers/chemistry
18.
Biomaterials ; 29(6): 733-41, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17997482

ABSTRACT

Sustained release of functional plasmid DNA from the surfaces of materials which support cell adhesion for tissue formation could have a significant impact on gene therapy and tissue engineering. We report here layer-by-layer assembled multilayer film from a degradable cationic poly(2-aminoethyl propylene phosphate) and plasmid DNA encoding for enhanced green fluorescent protein (EGFP) for mouse osteoblast cell adhesion and prolonged gene delivery. Multilayer film growth was monitored by UV spectrophotometry and intensity of absorbance at 260 nm related to incorporated DNA increased in an exponential manner with increase the number of deposited polymer and plasmid layers. It degraded upon incubation in phosphate-buffered saline (PBS) at 37 degrees C and sustained the release of bioactive plasmid DNA up to 2 months. The multilayer film facilitated initial mouse osteoblast cell adhesion onto the surface and enhanced cellular alkaline phosphatase activity and calcium accumulation. It sustained delivering transcriptional active DNA to mouse osteoblast cells cultured on the film, and directly prolonged gene expression in the presence of serum without any exogenous transfection agent. This biodegradable multilayer assembly is promising for the local and sustained delivery of plasmid DNA and such a layer-by-layer system suggests an alternative method for plasmid DNA incorporation which may be useful for surface modification of implanted materials or scaffold for gene therapy and tissue regeneration.


Subject(s)
Cations/metabolism , DNA/metabolism , Osteoblasts/metabolism , Plasmids , Animals , Mice , Spectrophotometry, Ultraviolet
19.
Biomacromolecules ; 8(11): 3375-81, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17902689

ABSTRACT

Novel biodegradable hydrogels by photo-cross-linking macromers based on polyphosphoesters and poly(ethylene glycol) (PEG) are reported. Photo-cross-linkable macromers were synthesized by ring-opening polymerization of the cyclic phosphoester monomer 2-(2-oxo-1,3,2-dioxaphospholoyloxy) ethyl methacrylate (OPEMA) using PEG as the initiator and stannous octoate as the catalyst. The macromers were characterized by 1H NMR, Fourier transform infrared spectroscopy, and gel permeation chromatography measurements. The content of polyphosphoester in the macromer was controlled by varying the feed ratio of OPEMA to PEG. Hydrogels were fabricated by exposing aqueous solutions of macromers with 0.05% (w/w) photoinitiator to UV light irradiation, and their swelling kinetics as well as degradation behaviors were evaluated. The results demonstrated that cross-linking density and pH values strongly affected the degradation rates. The macromers was compatible to osteoblast cells, not exhibiting significant cytotoxicity up to 0.5 mg/mL. "Live/dead" cell staining assay also demonstrated that a large majority of the osteoblast cells remained viable after encapsulation into the hydrogel constructs, showing their potential as tissue engineering scaffolds.


Subject(s)
Esters/chemistry , Hydrogels/chemical synthesis , Phosphorus/chemistry , Polyethylene Glycols/chemistry , Biodegradation, Environmental , Cell Survival/drug effects , Cells, Cultured , Esters/chemical synthesis , Esters/toxicity , Hydrogels/chemistry , Hydrogels/toxicity , Magnetic Resonance Spectroscopy , Microscopy, Electron, Scanning , Molecular Structure , Molecular Weight , Osteoclasts/cytology , Osteoclasts/drug effects , Phosphorus/toxicity , Photochemistry , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/toxicity , Spectroscopy, Fourier Transform Infrared
SELECTION OF CITATIONS
SEARCH DETAIL
...