Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Clin Hypertens ; 28(1): 40, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36451242

ABSTRACT

BACKGROUND: To assess the efficacy and safety of a combination therapy involving fimasartan, amlodipine, and rosuvastatin in patients with essential hypertension and dyslipidemia who fail to respond to fimasartan monotherapy. METHODS: This phase III, randomized, double-blind, multicenter study was conducted in adults aged 19-70 years. Patients who voluntarily consented were screened for eligibility to enroll in the study. Patients who failed to respond to 4 weeks of fimasartan monotherapy were randomized with a 1:1:1 ratio to the fimasartan 60 mg/amlodipine 10 mg + rosuvastatin 20 mg (FMS/ALD + RSV) as study group, fimasartan 60 mg/amlodipine 10 mg (FMS/ALD) as control 1 group, and fimasartan 60 mg + rosuvastatin 20 mg (FMS + RSV) as control 2 group. The primary efficacy endpoints were the change in the sitting systolic blood pressure and the rate of change in the low-density lipoprotein cholesterol (LDL-C) level from baseline to 8 weeks. The adverse events, adverse drug reactions, physical examination findings, laboratory test results, electrocardiograms, and vital signs were evaluated to assess safety in the study. RESULTS: Of 138 randomized patients, 131 were conducted efficacy analysis, and 125 completed the study. For the change in LDL-C and sitting SBP (SiSBP) as primary efficacy assessments, the change in LDL-C at week 8 was significantly reduce in the FMS/ALD + RSV group than in the control 1 group (P < 0.001). The change in SiSBP at week 8 were greater reduce in the FMS/ALD + RSV group than in the FMS + RSV group (both P < 0.001). For the safety evaluation, there were no differences among the treatment groups in the incidence of adverse drug reactions. CONCLUSIONS: The fimasartan/amlodipine + rosuvastatin combination therapy can effectively and safely lower blood pressure and improve lipid levels in patients with essential hypertension and dyslipidemia who fail to respond adequately to fimasartan monotherapy. TRIAL REGISTRATION: NCT03156842, Registered 17 May 2017.

2.
Clin Hypertens ; 24: 10, 2018.
Article in English | MEDLINE | ID: mdl-30186623

ABSTRACT

BACKGROUND: Fimasartan (Kanarb; Boryung Pharmaceutical Co., Ltd., Seoul, Republic of Korea) is a non-protein angiotensin II receptor blocker that selectively blocks the AT1 receptor. No prior large-scale study has investigated the impact of demographics, disease, treatment, and clinical characteristics on medication satisfaction and quality of life in Korean hypertensive patients. Additionally, it is unclear whether increased medication compliance affects the achievement of hypertension treatment objectives. METHODS: This was a multicenter, non-interventional, open-label and 8-week switching study. This study was divided into 2 steps. STEP I was a cross-sectional study composed entirely of hypertensive patients undergoing treatment and STEP II was a prospective observational study of hypertensive patients switching to fimasartan. A total of 12,244 and 2023 patients were analyzed in the STEP I and STEP II groups, respectively. In STEP I, we investigated demographics, clinical, disease, and treatment characteristics at the registration point and then analyzed medication satisfaction, patient compliance, and quality of life. In STEP II, the patients who switched to fimasartan were followed up for 8 weeks, and the data analyzed included changes in medication effects, satisfaction, compliance, and adverse events. RESULTS: Some baseline characteristics, such as sex, body mass index, region of residence, educational level, and income level, affected the quality of life and medication duration in hypertensive patients. At 4 and 8 weeks, 62.5 and 69.9% of patients, respectively, reached their target blood pressure. The medication satisfaction scores were increased 4.0 ± 1.2, 5.1 ± 1.1, and 5.4 ± 1.0 at baseline, 4 weeks, and 8 weeks, respectively, and the difference was statistically significant (p < 0.0001). Most patients (76.4%) who changed from prior antihypertensive drug to fimasartan were not satisfied with conventional antihypertensive drugs (e.g., lack of efficacy). Among 2183 patients, 234 adverse events occurred in 151 (6.9%) and 50 adverse drug reactions occurred in 39 (1.8%). CONCLUSION: The demographic, clinical, disease, and treatment characteristics of hypertensive patients were investigated in this study. After switching to fimasartan, blood pressure was significantly decreased and patient satisfaction was improved. Fimasartan treatment was well tolerated and safe in hypertensive patients in Korea.Trial registration. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: (NCT02394392).

3.
Transl Clin Pharmacol ; 26(3): 118-127, 2018 Sep.
Article in English | MEDLINE | ID: mdl-32055561

ABSTRACT

The safety and efficacy of fimasartan have been evaluated through post-marketing surveillance in real world clinical practice. The multi-center, prospective, open-label and non-interventional study. A total of 3,945 patients (3,729 patients for safety assessment and 3,473 patients for efficacy assessment) were screened in patients with essential hypertension in 89 study centers from 9 September 2010 through 8 September 2016. Among the total patients, 2,893 patients (77.6%) were administered fimasartan for 24 weeks or longer and were classified as 'patients with long-term follow-up', and the additional safety and efficacy analysis were performed. The improvement was defined as systolic blood pressure (SBP) controlled to ≤ 140 mmHg or decreased SBP differences ≥ 20 mmHg after treatment or diastolic blood pressure (DBP) controlled to ≤ 90 mmHg or decreased DBP differences ≥ 10 mmHg after treatment. Adverse drug reactions (ADRs) were reported in 3.8% patients; dizziness, and hypotension were the most frequently reported ADRs in total patients. The results of patients with long-term follow-up were comparable with total patients. The overall improvement rate in all efficacy assessment at the last visit was 87.1% (3,025/3,473 patients). The overall improvement rate of the patients with long-term follow-up was 88.9%. Fimasartan was well tolerated, with no new safety concerns identified and an effective treatment in the real world clinical practice for Korean patients with hypertension.

4.
J Appl Toxicol ; 33(12): 1474-83, 2013 Dec.
Article in English | MEDLINE | ID: mdl-22987290

ABSTRACT

Human endogenous retrovirus (HERV) envelope protein-coated, baculovirus vector-based HPV 16 L1 (AcHERV-HPV16L1) is a non-replicating recombinant baculoviral vaccine. Here, we report an initial evaluation of the preclinical safety of AcHERV-HPV16L1 vaccine. In an acute toxicity study, a single administration of AcHERV-HPV16L1 DNA vaccine given intramuscularly (i.m.) to mice at a dose of 1 × 10(8) plaque-forming units (PFU) did not cause significant changes in body weight compared with vehicle-treated controls. It did cause a brief increase in the weights of some organs on day 15 post-treatment, but by day 30, all organ weights were not significantly different from those in the vehicle-treated control group. No hematological changes were observed on day 30 post-treatment. In a range-finding toxicity study with three doses of 1 × 10(7) , 2 × 10(7) and 5 × 10(7) PFU once daily for 5 days, the group treated with 5 × 10(7) PFU showed a transient decrease in the body weights from day 5 to day 15 post-treatment, but recovery to the levels similar to those in the vehicle-treated control group by post-treatment day 20. Organ weights were slightly higher for lymph nodes, spleen, thymus and liver after repeated dosing with 5 × 10(7) PFU on day 15, but had normalized by day 30. Moreover, repeated administration of AcHERV-HPV16L1 did not induce myosin-specific autoantibody in serum, and did not cause immune complex deposition or tissue damage at injection sites. Taken together, these results provide preliminary evidence of the preclinical safety of AcHERV-based HPV16L1 DNA vaccines in mice.


Subject(s)
Baculoviridae/genetics , Capsid Proteins/genetics , Endogenous Retroviruses/genetics , Oncogene Proteins, Viral/genetics , Papillomavirus Vaccines/toxicity , Vaccines, DNA/toxicity , Viral Envelope Proteins/genetics , Animals , Autoantibodies/blood , Body Weight/drug effects , Dose-Response Relationship, Drug , Female , Genetic Vectors , Humans , Mice , Mice, Inbred BALB C , Organ Size/drug effects , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Sf9 Cells , Spodoptera/virology , Toxicity Tests, Acute , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
5.
J Control Release ; 164(2): 213-20, 2012 Dec 10.
Article in English | MEDLINE | ID: mdl-22664473

ABSTRACT

Nanoparticles have demonstrated potential for promoting drug delivery to tumor sites and enhancing uptake. Here, we report tetraiodothyroacetic acid (tetrac) as a promising new targeting moiety for delivery of anticancer drugs to tumor tissues. Tetrac, an antagonist that blocks the binding of thyroid hormone to integrin αvß3, was covalently linked to the activated end of pegylated lipid and used to formulate tetrac-tagged pegylated liposomes (TPL). After incubating with TPL for 9h, cellular accumulation efficiency into A375 human melanoma cells, which express integrin αvß3 at high density, was high (98.5%± 0.5% of cells), whereas that in KB cells, which express integrin at a very low density, was much lower (35.1%± 4.5%). Molecular imaging revealed that TPL preferentially distributed to tumor tissues after systemic administration in mice, where as non-targeting pegylated liposomes were distributed to tumors at background levels. Treatment with the alkyl lysophospholipid anticancer drug edelfosine, encapsulated in TPL, significantly reduced the survival of A375 tumor cells compared to cells treated with edelfosine in pegylated liposomes or with lysophosphatidylcholine encapsulated in TPL. Moreover, intravenous administration of edelfosine in TPL significantly reduced the growth of tumors and prolonged the survival of A375-xenografted mice, providing 100% protection for up to 50 days and some protection until 66 days (0% survival endpoint). In contrast, no untreated mice or mice treated with edelfosine-loaded pegylated liposomes survived up to 50 or 48 days, respectively, after tumor inoculation. These results suggest the potential of tetrac as a new ligand moiety for enhancing the delivery of anticancer drug-loaded nanoparticles to tumors and enhancing the therapeutic efficacy of encapsulated anticancer drugs.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Integrin alphaVbeta3/metabolism , Neoplasms/metabolism , Phospholipid Ethers/administration & dosage , Thyroxine/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Liposomes , Mice , Mice, Nude , Neoplasms/drug therapy , Phospholipid Ethers/chemistry , Thyroxine/administration & dosage , Thyroxine/chemistry
6.
J Control Release ; 154(3): 214-7, 2011 Sep 25.
Article in English | MEDLINE | ID: mdl-21763738

ABSTRACT

When tumor cells undergo apoptosis in response to chemotherapy, the levels of apoptotic biomarkers such as histone H1 are increased at the tumor. This would amplify in situ homing signals and thus drug delivery by apoptosis-targeted drugs. To examine this possibility, we prepared apoptosis-targeted liposomes containing doxorubicin by labeling them with the CQRPPR peptide (ApoPep-1) that recognizes apoptotic cells by binding to histone H1. ApoPep-1-labeled liposomes, but not folate-labeled liposomes, inhibited tumor growth in mice more efficiently than untargeted liposomes, although in vitro cytotoxicities of those liposomes were similar. Fluorescence imaging signals at tumor were increased by the homing of ApoPep-1-labeled, fluorescent liposomes, which was correlated with the increase of apoptosis and the amount of doxorubicin at the tumor and, conversely, with the decrease of tumor volume. These results demonstrate that the apoptosis-targeted drug delivery enables in situ dose amplification and, when combined with imaging of apoptosis, provides a real-time monitoring of treatment response for cancer theragnosis.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Apoptosis/drug effects , Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Histones/metabolism , Liposomes/chemistry , Animals , Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/therapeutic use , Liposomes/metabolism , Mice , Neoplasms/drug therapy , Peptides/chemistry , Peptides/metabolism
7.
Biomaterials ; 32(20): 4621-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21440296

ABSTRACT

To improve vaccine delivery, we herein designed a co-delivery system using a protein antigen and its encoding plasmid linked in nanocomplexes via maltosylated PEI (mPEI). Cationic mPEI was electrostatically complexed to a plasmid encoding the human papillomavirus (HPV) type 16L1 protein (pHPV16L1), and further complexed to a maltose binding protein (MBP)-fused human papillomavirus type 16L1 fusion protein (HPV16L1-MBP). The HPV16L1-MBP/mPEI/pHPV16L1 complexes were characterized by gel-retardation properties, zeta potentials and sizes. The intracellular co-delivery of protein and plasmid DNA vaccines was significantly higher for mPEI-based triple nanocomplexes than for a simple physical mixture of the proteins and DNA. Moreover, the cellular delivery of plasmid DNA using mPEI-based triple nanocomplexes resulted in higher expression levels comparable to those obtained using dual complexes of mPEI and the plasmid DNA. In vivo, co-immunization of mice with HPV16L1-MBP/mPEI/pHPV16L1 nanocomplexes triggered the highest levels of humoral immune responses among various vaccination groups. Moreover, the mPEI-based nanocomplexes significantly enhanced the number of interferon-γ producing CD8(+) T cells compared with the use of mixed proteins and plasmid DNA. These results suggest that the effective cellular co-delivery of MBP-fused antigen proteins and plasmid DNA using maltosylated PEI-based triple nanocomplexes could enhance the immunogenicity of HPV16L1 vaccines.


Subject(s)
Capsid Proteins/immunology , DNA/chemistry , Maltose-Binding Proteins/immunology , Nanostructures/chemistry , Oncogene Proteins, Viral/immunology , Polyethyleneimine/chemistry , Recombinant Fusion Proteins/immunology , Vaccines, DNA , Animals , CD8-Positive T-Lymphocytes/immunology , Capsid Proteins/genetics , Cell Line , DNA/metabolism , Humans , Maltose-Binding Proteins/genetics , Materials Testing , Mice , Molecular Structure , Oncogene Proteins, Viral/genetics , Particle Size , Polyethyleneimine/metabolism , Recombinant Fusion Proteins/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
8.
Mol Pharm ; 8(2): 430-8, 2011 Apr 04.
Article in English | MEDLINE | ID: mdl-21222482

ABSTRACT

Targeted delivery of imaging agents and therapeutics to tumors would provide early detection and increased therapeutic efficacy against cancer. Here we have screened a phage-displayed peptide library to identify peptides that selectively bind to lung tumor cells. Evaluation of individual phage clones after screening revealed that a phage clone displaying the CSNIDARAC peptide bound to H460 lung tumor cells at higher extent than other phage clones. The synthetic CSNIDARAC peptide strongly bound to H460 cells and was efficiently internalized into the cells, while little binding of a control peptide was seen. It also preferentially bound to other lung tumor cell lines as compared to cells of different tumor types. In vivo imaging of lung tumor was achieved by homing of fluorescence dye-labeled CSNIDARAC peptide to the tumor after intravenous injection into mice. Ex vivo imaging and microscopic analysis of isolated organs further demonstrated the targeting of CSNIDARAC peptide to tumor. The CSNIDARAC peptide-targeted and doxorubicin-loaded liposomes inhibited the tumor growth more efficiently than untargeted liposomes or free doxorubicin. In vivo imaging of fluorescence dye-labeled liposomes demonstrated selective homing of the CSNIDARAC-liposomes to tumor. In the same context, higher levels of doxorubicin and apoptosis in tumor tissue were observed when treated with the targeted liposomes than untargeted liposomes or free doxorubicin. These results suggest that the CSNIDARAC peptide is a promising targeting probe that is able to direct imaging agents and therapeutics to lung tumor.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Peptide Fragments/administration & dosage , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Blotting, Western , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , Immunoenzyme Techniques , Liposomes , Mice , Mice, Inbred BALB C , Mice, Nude , Peptide Fragments/pharmacokinetics , Peptide Library , Tissue Distribution , Tumor Cells, Cultured
9.
Biomaterials ; 32(3): 849-57, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20926129

ABSTRACT

Amphiphilic α-tocopherol oligochitosan conjugates were constructed by conjugating α-tocopherol succinate to water soluble oligochitosans with various molecular weights. In aqueous medium, the tocopherol oligochitosan conjugates self-assembled to single layered oligomersomes. The sizes of α-tocopherol-oligochitosan-based oligomersomes (TCOsomes) could be controlled by chain lengths of oligochitosans. The mean sizes of TCOsomes were 220 and 377 nm as the sizes of oligochitosans were 4000 and 12,500, respectively. For all TCOsomes formed in this study, polydispersity indexes were in the ranges of 0.111-0.256. Cryo-TEM images showed clear thickening in the unilamellar layer of TCOsomes upon complexation with siRNAs. Zeta potentials decreased as the ratios of siRNA/TCOsomes increased. TCOsomes self-assembled from tocopherol-oligochitosan 4K (TCOsome(4K)) significantly enhanced the cellular uptake of siRNAs (>98%), and reduced the expression of target proteins more effectively than did Lipofectamine 2000. In tumor xenografted mice, the intratumoral administration of siMcl-1 using TCOsomes substantially silenced the expression of Mcl-1 and prevented the growth of tumor. The hematoxylin-eosin staining showed the apoptosis of cells in the tissues of the mice treated with siMcl-1/TCOsome(4K) complexes, but not with siGL2/TCOsome(4K) complexes. The self-assembling and size-controllable oligomersomes might be suitable for effective in vivo delivery of siRNAs.


Subject(s)
Chitin/analogs & derivatives , Genetic Vectors/chemistry , RNA, Small Interfering/administration & dosage , Tocopherols/chemistry , Animals , Chitin/chemistry , Chitosan , Cryoelectron Microscopy , Female , Genetic Vectors/administration & dosage , Genetic Vectors/ultrastructure , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Transmission , Oligosaccharides , RNA, Small Interfering/chemistry , Xenograft Model Antitumor Assays
10.
J Control Release ; 155(1): 60-6, 2011 Oct 10.
Article in English | MEDLINE | ID: mdl-20971142

ABSTRACT

Oligolysine-based cationic lipid derivatives were synthesized for delivery of siRNA, and formulated into cationic liposomes. Among various oligolysine-based lipid derivatives differing in lysine residue number and lipid moiety, trilysinoyl oleylamide (TLO)-based liposomes (TLOL) showed the highest delivery efficiency combined with minimal cytotoxicity. Delivery of siRNA using TLOL silenced target genes both in vitro and in vivo. In green fluorescent protein (GFP)-expressing tumor tissue, a significant reduction of fluorescence was observed after intratumoral administration of siGFP using TLOL compared with control siGL2. Intravenous administration of siMcl1 employing pegylated TLOL (pTLOL) reduced the expression of human Mcl1 protein in KB-xenografted tumor tissue. Despite the reduction in target protein Mcl1 expression following such systemic delivery, tumor growth was only slightly reduced compared to a siGL2-treated control group. To potentiate the anticancer activity of siMcl1, the anticancer drug suberoylanilide hydroxamic acid (SAHA) was additionally encapsulated in pTLOL. After intravenous administration of siMcl1 using SAHA-loaded pTLOL (pSTLOL), a significant reduction in tumor growth was observed compared to that seen in animals treated with free SAHA or siGL2 complexed with pSTLOL. The results indicate that pTLOL could be further developed as a systemic delivery system for synergistic anticancer siRNA and a drug.


Subject(s)
Antineoplastic Agents/administration & dosage , Cations/chemistry , Hydroxamic Acids/administration & dosage , Liposomes/chemistry , Neoplasms/drug therapy , Oleic Acids/chemistry , RNA, Small Interfering/administration & dosage , Animals , Antineoplastic Agents/therapeutic use , Cell Line , Female , Gene Silencing , Humans , Hydroxamic Acids/therapeutic use , Mice , Mice, Nude , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasms/genetics , Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Vorinostat
11.
Nanomedicine ; 6(6): 769-76, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20570638

ABSTRACT

Intraperitoneal chemotherapy confers limited clinical benefit as a result of the dose-limiting toxicity of anticancer drugs. We aimed to develop optimized liposomes for mitoxantrone (MTO) administration that provide high encapsulation efficiency and increase the therapeutic index. Cationic MTO was loaded onto anionic liposomes by electrostatic surface complexation. The anticancer activity was evaluated in a peritoneal carcinomatosis model. The retention of MTO at the tumor site was monitored by molecular imaging. MTO loading efficiencies by electrostatic complexation were >95% for all anionic liposomes but <5% for neutral liposomes. Among anionic liposomes, cardiolipin liposomes (CLs) exhibited the strongest binding affinity for MTO, the highest anticancer activity, and the lowest toxicity. MTO delivered by CLs showed prolonged retention at tumor sites. Unlike free MTO showing significant cardiotoxicity, MTO administered in CLs provided negligible cardiotoxicity. CL-mediated delivery may increase the therapeutic index of MTO chemotherapy by prolonged retention and reduced cardiotoxicity.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Cardiolipins/chemistry , Liposomes/chemistry , Mitoxantrone/adverse effects , Mitoxantrone/chemistry , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Female , Injections, Intraperitoneal , Liposomes/administration & dosage , Mice , Mitoxantrone/pharmacology , Mitoxantrone/therapeutic use
12.
J Control Release ; 145(2): 159-64, 2010 Jul 14.
Article in English | MEDLINE | ID: mdl-20385182

ABSTRACT

For delivery of siRNA, chitosan (CS) was derivatized with poly-l-arginine (PLR) and polyethylene glycol (PEG). The formation of polyplexes with siRNA was confirmed by gel retardation. The PLR-grafted CS formed nanosized particles with siRNA. PLR-grafted CS showed higher cellular delivery efficiency of siRNA than did CS, pegylated CS, PLR, or pegylated PLR. The extent of reduction in the expression of fluorescent proteins was highest following treatment of the cells using PLR derivatives of CS in complexes with specific siRNAs. Cell viability was greater in populations treated with pegylated CS-PLR than in those treated with PLR. Hemolysis of erythrocytes was reduced upon conjugation of PLR with CS. The delivery of siRNAs via pegylated CS-PLR revealed little dependence on serum. Molecular imaging techniques revealed that the intratumoral administration of red fluorescent protein-specific siRNA in complexes with pegylated CS-PLR significantly silenced the expression of red fluorescent proteins in tumor tissues in vivo. These results indicate that pegylated CS-PLR might be useful for in vivo delivery of therapeutic siRNAs.


Subject(s)
Chitosan/chemistry , RNA, Small Interfering/administration & dosage , Animals , Arginine/chemistry , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers , Hemolysis/drug effects , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Mice , Molecular Weight , Polyethylene Glycols/chemistry , Polymers/chemistry , Polymers/pharmacology , Polymers/toxicity
13.
J Control Release ; 140(3): 268-76, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-19567256

ABSTRACT

Viable siRNA therapeutic strategies require the concurrent development of effective and safe delivery systems. Here, we described the synthesis of a new cationic lipid, N,N''-dioleylglutamide (DG), and evaluated DG-based liposomes as an siRNA delivery system. DG, an amino acid derivative, was synthesized by peptide bond linkage of oleylamine to each carboxylic acid group of glutamic acid. Gel retardation assays showed that DG-based cationic liposomes and siRNA began to form complexes from the N/P ratio of 1.8. The viability of A549, HeLa and WM266.4 cells was significantly higher after treatment with DG-based liposomes than with Lipofectamine 2000 and cationic 3beta-[N-(N',N'-dimethylaminoethane)-carbamoyl] cholesterol (DC-Chol)-based liposomes. The DG-based cationic liposomes could effectively deliver a fluorescent model siRNA into A549, HeLa, and WM266.4 human cancer cell lines, showing at least 2-fold higher fluorescence mean intensity values than did Lipofectamine 2000. When survivin-specific siRNA was delivered to cells in lipoplexes, survivin mRNA levels were reduced by DG-based liposomes to the higher extent than Lipofectamine 2000 and DC-Chol-based liposomes. When red fluorescent protein (RFP)-expressing cells were treated with RFP-specific siRNA (siRFP), RFP expression significantly decreased in cells treated with DG-based liposomes. Molecular imaging revealed that intratumoral injection of siRFP and DG-based liposome complexes significantly reduced fluorescence in RFP-expressing tumor tissues in mice. These results suggest that DG-based cationic liposomes would be of value for cellular delivery and in vivo local delivery of siRNA.


Subject(s)
Amino Acids/chemistry , Gene Silencing/drug effects , Glutamine/analogs & derivatives , Lipids/chemistry , Lipopeptides/chemistry , RNA, Small Interfering/administration & dosage , Animals , Cations/chemistry , Cell Line, Tumor , Chemistry, Pharmaceutical , Cholesterol/chemistry , Chromatography, Gel , Drug Carriers , Drug Delivery Systems , Electrochemistry , Glutamine/chemistry , Humans , Liposomes , Magnetic Resonance Spectroscopy , Mice , Particle Size , Phosphatidylethanolamines/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spectrophotometry, Ultraviolet
14.
J Drug Target ; 17(2): 123-32, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19012052

ABSTRACT

In this study, we tested the use of cationic polymer derivatives of biocompatible hyaluronic acid (HA) as a delivery system of siRNA and antisense oligonucleotides. HA was modified with cationic polymer polyethylenimine (PEI). When compared with PEI alone, cationic PEI derivatives of HA (HA-PEI) provided increased cellular delivery of Small interfering RNA (siRNA) in B16F1, A549, HeLa, and Hep3B tumor cells. Indeed, more than 95% of the cells were positive for siRNA following its delivery with HA-PEI. A survivin-specific siRNA that was delivered using HA-PEI potently reduced the mRNA expression levels of the target gene in all of the cell lines. By contrast, survivin-specific siRNA delivered by PEI alone did not induce a significant reduction in mRNA levels. In green fluorescent protein (GFP)-expressing 293 T cells, a loss of GFP expression was evident in the cells that had been treated with GFP-specific siRNA and HA-PEI complex. The inhibition of target gene expression by antisense oligonucleotide G3139 was also enhanced after delivery with HA-PEI. Moreover, HA-PEI displayed lower cytotoxicity than PEI alone. These results suggest that HA-PEI could be further developed as biocompatible delivery systems of siRNA and antisense oligonucleotides for enhanced cellular uptake and inhibition of target gene expression.


Subject(s)
Hyaluronic Acid/chemistry , Oligonucleotides, Antisense/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Cations , Cell Line, Tumor , Drug Carriers , Green Fluorescent Proteins/chemistry , Humans , Magnetic Resonance Spectroscopy , Oligonucleotides, Antisense/pharmacokinetics , Oligonucleotides, Antisense/pharmacology , Polyethyleneimine/chemistry , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology
15.
Biopolymers ; 89(7): 635-42, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18322932

ABSTRACT

A novel target specific small interfering RNA (siRNA) delivery system was successfully developed using polyethyleneimine (PEI)-hyaluronic acid (HA) conjugate. Anti-PGL3-Luc siRNA was used as a model system suppressing the PGL3-Luc gene expression. The siRNA/PEI-HA complex with an average size of ca. 21 nm appeared to be formed by electrostatic interaction between the negatively charged siRNA and the positively charged PEI of PEI-HA conjugate. The cytotoxicity of siRNA/PEI-HA complex to B16F1 cells was lower than that of siRNA/PEI complex according to the MTT assay. When B16F1 and HEK-293 cells were treated with fluorescein isothiocyanate (FITC) labeled siRNA/PEI-HA complex, B16F1 cells, with a lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1), showed higher green fluorescent intensity than HEK-293 cells because of the HA receptor mediated endocytosis of the complex. Accordingly, the PGL3-Luc gene silencing of anti-PGL3-Luc siRNA/PEI-HA complex was more efficient in B16F1 cells than in HEK-293 cells. In addition, the inhibited PGL3-Luc gene silencing effect in the presence of free HA in the transfection medium revealed that siRNA/HA-PEI complex was selectively taken up to B16F1 cells via HA receptor mediated endocytosis. All these results demonstrated that the intracellular delivery of anti-PGL3-Luc siRNA/PEI-HA complex could be facilitated by the HA receptor mediated endocytosis.


Subject(s)
Gene Transfer Techniques , Hyaluronic Acid/chemistry , Polyethyleneimine/chemistry , RNA, Small Interfering/chemistry , Cell Line , Fluorescein-5-isothiocyanate/chemistry , Gene Expression/drug effects , Magnetic Resonance Spectroscopy , Microscopy, Atomic Force , Molecular Structure , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology
16.
Int J Pharm ; 353(1-2): 260-9, 2008 Apr 02.
Article in English | MEDLINE | ID: mdl-18178348

ABSTRACT

Most cationic liposomes used for gene delivery suffer from reduced transfection efficiency in the presence of serum. In this study, we report serum-enhanced delivery efficiency of siRNA via the use of newly synthesized liposomes that contain cationic lipids. Two cholesterol derivatives, cholesteryloxypropan-1-amine (COPA) and cholesteryl-2-aminoethylcarbamate (CAEC), were synthesized. A fluorescein label was then used to visualize cellular uptake of small interfering RNA (siRNA) via COPA or CAEC-based liposomes. The presence of serum had different effects on the cellular delivery of siRNA when siRNA was complexed to different cationic liposomes. CAEC-based liposomes showed significantly reduced cellular delivery of siRNA in serum-containing media as compared to serum-free media. Conversely, COPA-based liposomes (COPA-L) provided serum-enhanced delivery of siRNA in Hepa1-6, A549, and Hela cell lines. Following delivery of the oncogene survivin-specific siRNA, COPA-L reduced the mRNA expression levels of the target gene more efficiently than did Lipofectamine 2000. The delivery of green fluorescent protein-specific siRNA with COPA-L reduced the expression of green fluorescent protein in 293T stable cell lines. The apoptosis of Hepa1-6 significantly increased by delivery of survivin-specific siRNA by COPA-L. Additionally, Hepa1-6, A549, and Hela cells were >80% viable after treatment with COPA-L. These results suggest that the newly synthesized cholesterol derivative, COPA-L, could be further developed as a serum-enhanced delivery system of siRNA.


Subject(s)
Cholesterol/chemistry , Liposomes , RNA, Small Interfering/administration & dosage , Serum , Apoptosis , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inhibitor of Apoptosis Proteins , Magnetic Resonance Spectroscopy , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Survivin
SELECTION OF CITATIONS
SEARCH DETAIL
...