2001
|
da Silva CL, Del Ciampo JO, Rossetti FC, Bentley MVLB, Pierre MBR. ImprovedIn vitroandIn vivoCutaneous Delivery of Protoporphyrin IX from PLGA-based Nanoparticles. Photochem Photobiol 2013; 89:1176-84. [DOI: 10.1111/php.12121] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 06/17/2013] [Indexed: 11/29/2022]
Affiliation(s)
- Carolina L. da Silva
- School of Pharmacy; Federal University of Rio de Janeiro; Rio de Janeiro; SP; Brazil
| | - José O. Del Ciampo
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo; Ribeirão Preto; SP; Brazil
| | - Fábia C. Rossetti
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo; Ribeirão Preto; SP; Brazil
| | - Maria V. L. B. Bentley
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo; Ribeirão Preto; SP; Brazil
| | - Maria B. R. Pierre
- School of Pharmacy; Federal University of Rio de Janeiro; Rio de Janeiro; SP; Brazil
| |
Collapse
|
2002
|
Razmi M, Divsalar A, Saboury AA, Izadi Z, Haertlé T, Mansuri-Torshizi H. Beta-casein and its complexes with chitosan as nanovehicles for delivery of a platinum anticancer drug. Colloids Surf B Biointerfaces 2013; 112:362-7. [PMID: 24028849 DOI: 10.1016/j.colsurfb.2013.08.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/15/2013] [Accepted: 08/18/2013] [Indexed: 12/25/2022]
Abstract
The clinical application of platinum-based anticancer drugs is greatly limited by severe toxicity. Drug-delivery systems are much sought after to improve the efficacy and applicability of these drugs. Here, we describe a new drug-delivery system comprising a novel platinum complex (bipyridine morpholine dithiocarbamate Pt(II) nitrate) within nanoparticles composed of β-casein (β-CN) and chitosan (CS). The influence of pH on the formation of a colloidally-stable nanocarrier system composed of Pt complex-loaded β-CN and chitosan nanoparticles was investigated using UV-vis spectrometry, dynamic light scattering (DLS) and scanning electron microscopy (SEM). The particles of Pt complex-loaded beta-casein-chitosan formed were stable and soluble in the pH range 5.7-6.2. Hence, the optimal pH for complex formation is between the pI of casein (5.3) and the pKa of chitosan (6.5). DLS data showed that, at both pH values of 5.7 and 6.2, the particles formed had sizes between 200 and 300nm. However, the optimum pH for particle formation was pH 5.7. At this pH, the zeta-potential values of nanoparticles were positive and the particles were stable. SEM analysis confirmed the formation of nanoparticles with good colloidal stability and an average particle size of 200nm. The cytotoxicity of both free and encapsulated Pt complex was evaluated on colorectal carcinoma HCT116 cells. The results obtained indicated that both the cytotoxicity and cellular uptake of platinum were enhanced by its entrapment in β-CN-CS nanovehicles. These findings suggest that this novel drug-delivery system enables drugs to be thermodynamically stable in aqueous solutions and is potentially useful for targeted oral-delivery applications.
Collapse
Affiliation(s)
- Mahdieh Razmi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
2003
|
Singh S, Singh AN, Verma A, Dubey VK. Biodegradable Polycaprolactone (PCL) Nanosphere Encapsulating Superoxide Dismutase and Catalase Enzymes. Appl Biochem Biotechnol 2013; 171:1545-58. [DOI: 10.1007/s12010-013-0427-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/05/2013] [Indexed: 11/30/2022]
|
2004
|
Huang SS, Li IH, Hong PD, Yeh MK. Evaluation of protective efficacy using a nonstructural protein NS1 in DNA vaccine-loaded microspheres against dengue 2 virus. Int J Nanomedicine 2013; 8:3161-9. [PMID: 23990724 PMCID: PMC3753149 DOI: 10.2147/ijn.s49972] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dengue virus results in dengue fever or severe dengue hemorrhagic fever/dengue shock syndrome in humans. The purpose of this work was to develop an effective antidengue virus delivery system, by designing poly (dl-lactic-co-glycolic) acid/polyethylene glycol (PLGA/PEG) microspheres using a double-emulsion solvent extraction method, for vaccination therapy based on locally and continuously sustained biological activity. Nonstructural protein 1 (NS1) in deoxyribonucleic acid (DNA) vaccine–loaded PLGA/PEG microspheres exhibited a high loading capacity (4.5% w/w), yield (85.2%), and entrapment efficiency (39%), the mean particle size 4.8 μm, and a controlled in vitro release profile with a low initial burst (18.5%), lag time (4 days), and continued released protein over 70 days. The distribution of protein on the microspheres surface, outer layer, and core were 3.0%, 28.5%, and 60.7%, respectively. A release rate was noticed to be 1.07 μg protein/mg microspheres/day of protein release, maintained for 42 days. The cumulative release amount at Days 1, 28, and 42 was 18.5, 53.7, and 62.66 μg protein/mg microspheres, respectively. The dengue virus challenge in mice test, in which mice received one dose of 20 μg NS1 protein content of microspheres, in comparison with NS1 protein in Al(OH)3 or PBS solution, was evaluated after intramuscular immunization of BALB/c mice. The study results show that the greatest survival was observed in the group of mice immunized with NS1 protein–loaded PLGA/PEG microspheres (100%). In vivo vaccination studies also demonstrated that NS1 protein–loaded PLGA/PEG microspheres had a protective ability; its steady-state immune protection in rat plasma changed from 4,443 ± 1,384 pg/mL to 10,697 ± 3,197 pg/mL, which was 2.5-fold higher than that observed for dengue virus in Al(OH)3 at 21 days. These findings strongly suggest that NS1 protein–loaded PLGA/PEG microspheres offer a new therapeutic strategy in optimizing the vaccine incorporation and delivery properties of these potential vaccine targeting carriers.
Collapse
Affiliation(s)
- Shih-shiung Huang
- Biomedical Engineering Program, graduate Institute of Applied Science and Technology, and Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taiwan
| | | | | | | |
Collapse
|
2005
|
Qiu JF, Gao X, Wang BL, Wei XW, Gou ML, Men K, Liu XY, Guo G, Qian ZY, Huang MJ. Preparation and characterization of monomethoxy poly(ethylene glycol)-poly(ε-caprolactone) micelles for the solubilization and in vivo delivery of luteolin. Int J Nanomedicine 2013; 8:3061-9. [PMID: 23990719 PMCID: PMC3748903 DOI: 10.2147/ijn.s45062] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Luteolin (Lu) is one of the flavonoids with anticancer activity, but its poor water solubility limits its use clinically. In this work, we used monomethoxy poly(ethylene glycol)-poly(e-caprolactone) (MPEG-PCL) micelles to encapsulate Lu by a self-assembly method, creating a water-soluble Lu/MPEG-PCL micelle. These micelles had a mean particle size of 38.6 ± 0.6 nm (polydispersity index = 0.16 ± 0.02), encapsulation efficiency of 98.32% ± 1.12%, and drug loading of 3.93% ± 0.25%. Lu/MPEG-PCL micelles could slowly release Lu in vitro. Encapsulation of Lu in MPEG-PCL micelles improved the half-life (t½ ; 152.25 ± 49.92 versus [vs] 7.16 ± 1.23 minutes, P = 0.007), area under the curve (0-t) (2914.05 ± 445.17 vs 502.65 ± 140.12 mg/L/minute, P = 0.001), area under the curve (0-∞) (2989.03 ± 433.22 vs 503.81 ± 141.41 mg/L/minute, P = 0.001), and peak concentration (92.70 ± 11.61 vs 38.98 ± 7.73 mg/L, P = 0.003) of Lu when the drug was intravenously administered at a dose of 30 mg/kg in rats. Also, Lu/MPEG-PCL micelles maintained the cytotoxicity of Lu on 4T1 breast cancer cells (IC50 = 6.4 ± 2.30 μg/mL) and C-26 colon carcinoma cells (IC50 = 12.62 ± 2.17 μg/mL) in vitro. These data suggested that encapsulation of Lu into MPEG-PCL micelles created an aqueous formulation of Lu with potential anticancer effect.
Collapse
Affiliation(s)
- Jin-Feng Qiu
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and Medical School, Sichuan University, Chengdu, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
2006
|
Weiss VU, Lehner A, Grombe R, Marchetti-Deschmann M, Allmaier G. Chip electrophoresis of gelatin-based nanoparticles. Electrophoresis 2013; 34:2152-61. [DOI: 10.1002/elps.201300074] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 04/30/2013] [Accepted: 05/01/2013] [Indexed: 01/10/2023]
Affiliation(s)
- Victor U. Weiss
- Institute of Chemical Technologies and Analytics; Vienna University of Technology; Vienna; Austria
| | - Angela Lehner
- Institute of Chemical Technologies and Analytics; Vienna University of Technology; Vienna; Austria
| | - Ringo Grombe
- EC - Joint Research Centre - Institute for Reference Materials and Measurements; Geel; Belgium
| | | | - Günter Allmaier
- Institute of Chemical Technologies and Analytics; Vienna University of Technology; Vienna; Austria
| |
Collapse
|
2007
|
de Souza JRR, Feitosa JP, Ricardo NM, Trevisan MTS, de Paula HCB, Ulrich CM, Owen RW. Spray-drying encapsulation of mangiferin using natural polymers. Food Hydrocoll 2013. [DOI: 10.1016/j.foodhyd.2013.02.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
2008
|
Comparative evaluation of novel biodegradable nanoparticles for the drug targeting to breast cancer cells. Eur J Pharm Biopharm 2013; 85:463-72. [PMID: 23916461 DOI: 10.1016/j.ejpb.2013.07.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 07/18/2013] [Accepted: 07/23/2013] [Indexed: 11/20/2022]
Abstract
Nanomedicine formulations such as biodegradable nanoparticles (nps) and liposomes offer several advantages over traditional routes of administration: due to their small size, nanocarriers are able to selectively accumulate inside tumours or inflammatory tissues, resulting in improved drug efficacy and reduced side effects. To further augment targeting ability of nanoparticles towards tumour cells, specific ligands or antibodies that selectively recognise biomarkers over-expressed on cancer cells, can be attached to the surface either by chemical bond or by hydrophilic/hydrophobic interactions. In the present work, Herceptin (HER), a monoclonal antibody (mAb) able to selectively recognise HER-2 over-expressing tumour cells (such as breast and ovarian cancer cells), was absorbed on the surface of nanoparticles through hydrophilic/hydrophobic interactions. Nps were prepared by a modified single emulsion solvent evaporation method with five different polymers: three commercial polyesters (poly(ε-caprolactone) (PCL), poly (D,L-lactide) (PLA) and poly (D,L-lactide-co-.glycolide) (PLGA)) and two novel biodegradable polyesterurethanes (PURs) based on Poly(ε-caprolactone) blocks, synthesised with different chain extenders (1,4-cyclohexane dimethanol (CDM) and N-Boc-serinol). Polyurethanes were introduced as matrix-forming materials for nanoparticles due to their high chemical versatility, which allows tailoring of the materials final properties by properly selecting the reagents. All nps exhibited a small size and negative surface charge, suitable for surface functionalisation with mAb through hydrophilic/hydrophobic interactions. The extent of cellular internalisation was tested on two different cell lines: MCF-7 and SK-BR-3 breast cancer cells showing a normal and a high expression of the HER-2 receptor, respectively. Paclitaxel, a model anti-neoplastic drug, was encapsulated inside all nps, and release profiles and cytotoxicity on SK-BR-3 cells were also assessed. Interestingly, PUR nps were superior to commercial polyester-based nps in terms of higher cellular internalisation and cytotoxic activity on the tested cell lines. Results obtained warrants further investigation on the application of these PUR nps for controlled drug delivery and targeting.
Collapse
|
2009
|
Hao S, Wang B, Wang Y, Zhu L, Wang B, Guo T. Preparation of Eudragit L 100-55 enteric nanoparticles by a novel emulsion diffusion method. Colloids Surf B Biointerfaces 2013; 108:127-33. [DOI: 10.1016/j.colsurfb.2013.02.036] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 02/21/2013] [Accepted: 02/22/2013] [Indexed: 10/27/2022]
|
2010
|
Bastakoti BP, Liao SH, Inoue M, Yusa SI, Imura M, Nakashima K, Wu KCW, Yamauchi Y. pH-responsive polymeric micelles with core-shell-corona architectures as intracellular anti-cancer drug carriers. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2013; 14:044402. [PMID: 27877587 PMCID: PMC5090313 DOI: 10.1088/1468-6996/14/4/044402] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 06/03/2013] [Indexed: 06/04/2023]
Abstract
Polymeric micelles with core-shell-corona nanoarchitecture were designed for intracellular therapeutic anti-cancer drug carriers. Poly(styrene-b-acrylic acid-b-ethylene glycol) (PS-b-PAA-b-PEG) asymmetric triblock copolymer underwent self-assembly in aqueous solution to form spherical micelles with hydrophobic PS core, anionic PAA shell and hydrophilic PEG corona. The anti-cancer drug (doxorubicin, DOX) was successfully incorporated into the polymeric micelles. The in vitro release experiment confirmed that the release of DOX from the micelles was inhibited at pH 7.4. In contrast, an accelerated release of DOX was observed at mildly acidic conditions such as pH 4.5. The excellent biocompatibility of our PS-b-PAA-b-PEG-based micelles made the synthesized nano-carrier best suited for the delivery of anti-cancer drugs.
Collapse
Affiliation(s)
- Bishnu Prasad Bastakoti
- World Premier International (WPI) Research Center for Materials, Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Shih-Hsiang Liao
- Department of Chemical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Masamichi Inoue
- Department of Materials Science and Chemistry, University of Hyogo, 2167 Shosha, Himeji 671-2280, Japan
| | - Shin-Ichi Yusa
- Department of Materials Science and Chemistry, University of Hyogo, 2167 Shosha, Himeji 671-2280, Japan
| | - Masataka Imura
- World Premier International (WPI) Research Center for Materials, Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Kenichi Nakashima
- Department of Chemistry, Faculty of Science and Engineering, Saga University, 1 Honjo-machi, Saga 840-8502, Japan
| | - Kevin C-W Wu
- Department of Chemical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
- Division of Medical Engineering Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 350, Taiwan
| | - Yusuke Yamauchi
- World Premier International (WPI) Research Center for Materials, Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
2011
|
VEGF-releasing biodegradable nanospheres administered by craniotomy: A novel therapeutic approach in the APP/Ps1 mouse model of Alzheimer's disease. J Control Release 2013; 170:111-9. [DOI: 10.1016/j.jconrel.2013.04.028] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 04/25/2013] [Accepted: 04/27/2013] [Indexed: 11/17/2022]
|
2012
|
Rossi F, Ferrari R, Papa S, Moscatelli D, Casalini T, Forloni G, Perale G, Veglianese P. Tunable hydrogel—Nanoparticles release system for sustained combination therapies in the spinal cord. Colloids Surf B Biointerfaces 2013; 108:169-77. [DOI: 10.1016/j.colsurfb.2013.02.046] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 02/26/2013] [Accepted: 02/27/2013] [Indexed: 01/25/2023]
|
2013
|
Swet JH, Pacheco HJ, Iannitti DA, El-Ghanam A, McKillop IH. A silica-calcium-phosphate nanocomposite drug delivery system for the treatment of hepatocellular carcinoma: in vivo study. J Biomed Mater Res B Appl Biomater 2013; 102:190-202. [PMID: 23913418 DOI: 10.1002/jbm.b.32995] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 05/28/2013] [Accepted: 06/06/2013] [Indexed: 11/06/2022]
Abstract
Hepatocellular carcinoma (HCC) is notoriously difficult to treat with systemic chemotherapy. The aim of this study was to evaluate a silica-calcium-phosphate nanocomposite (SCPC75) drug delivery system (DDS) as a means to localize cisplatin treatment within the tumor, while reducing systemic toxicity, in a rat model of HCC. The SCPC75 was prepared and loaded with cisplatin and Fourier transform infrared analyses demonstrated even drug distribution within the SCPC75. A rat model of subcutaneous HCC formation was established and animals treated by either systemic cisplatin injection (sCis) or with SCPC75-Cis hybrid placed adjacent (ADJ) to or within (IT) the tumor. Five days after implantation, 50-55% of the total cisplatin loaded had been released from the SCPC75-Cis hybrids resulting in an approximately 50% decrease in tumor volume compared with sCis treatment. sCis-treated animals exhibited severe side effects, including rapid weight loss and decreased liver and kidney function, effects not observed in SCPC75-Cis-treated animals. Analysis of cisplatin distribution demonstrated drug concentrations in the tumor were 21 and 1.5 times higher in IT and ADJ groups, respectively, compared with sCis-treated animals. These data demonstrate the SCPC75 DDS can provide an effective, localized treatment for HCC with significantly reduced toxicity when compared with systemic drug administration.
Collapse
Affiliation(s)
- Jacob H Swet
- Department of Surgery, Carolinas Medical Center, Charlotte, North Carolina, 28203
| | | | | | | | | |
Collapse
|
2014
|
Thanki K, Gangwal RP, Sangamwar AT, Jain S. Oral delivery of anticancer drugs: Challenges and opportunities. J Control Release 2013; 170:15-40. [DOI: 10.1016/j.jconrel.2013.04.020] [Citation(s) in RCA: 330] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 04/25/2013] [Accepted: 04/26/2013] [Indexed: 12/12/2022]
|
2015
|
Nazarian S, Gargari SLM, Rasooli I, Hasannia S, Pirooznia N. A PLGA-encapsulated chimeric protein protects against adherence and toxicity of enterotoxigenic Escherichia coli. Microbiol Res 2013; 169:205-12. [PMID: 23906742 DOI: 10.1016/j.micres.2013.06.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 05/20/2013] [Accepted: 06/14/2013] [Indexed: 01/14/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) are the most common cause of diarrhea among children. Colonization factors and enterotoxins are the major ETEC candidate vaccines. Since protection against ETEC mostly occurs by induction of IgA antibodies, much effort is focused on the development of oral vaccines. In this study oral immunogenicity of a poly(lactic-co-glycolic acid) (PLGA) encapsulated chimeric protein containing CfaB, CstH, CotA and LTB (Heat-labile B subunit) was investigated. The protein was encapsulated in PLGA by double emulsion method and nanoparticles were characterized physicochemically. Immunogenicity was assessed by evaluating IgG1, IgG2 and IgA titers after BALB/c mice vaccination. Non aggregated nanoparticles had a spherical shape with an average particle size of 252.7±23 nm and 91.96±4.4% of encapsulation efficiency. Western blotting showed maintenance of the molecular weight and antigenicity of the released protein. Oral immunization of mice induced serum IgG and fecal IgA antibody responses. Immunization induced protection against ETEC binding to Caco-2 cells. The effect of LT toxin on fluid accumulation in ileal loops was neutralized by inhibition of enterotoxin binding to GM1-ganglosides. Delivery of the chimeric protein in PLGA elicited both systemic and mucosal immune responses. The findings could be exploited to development of oral multi-component ETEC prophylactic measures.
Collapse
Affiliation(s)
| | | | - Iraj Rasooli
- Department of Biology, Shahed University, Tehran, Iran
| | - Sadegh Hasannia
- Tarbiat Modares University, Faculty of Biological Sciences, Tehran, Iran
| | - Nazanin Pirooznia
- Faculty of Science, Department of Biology, University of Guilan, Rasht, Iran
| |
Collapse
|
2016
|
Yu Y, Tan S, Zhao S, Zhuang X, Song Q, Wang Y, Zhou Q, Zhang Z. Antitumor activity of docetaxel-loaded polymeric nanoparticles fabricated by Shirasu porous glass membrane-emulsification technique. Int J Nanomedicine 2013; 8:2641-52. [PMID: 23935362 PMCID: PMC3735276 DOI: 10.2147/ijn.s48214] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Docetaxel (DTX) has excellent efficiency against a wide spectrum of cancers. However, the current clinical formulation has limited its usage, as it causes some severe side effects. Various polymeric nanoparticles have thus been developed as alternative formulations of DTX, but they have been mostly fabricated on a laboratory scale. Previously, we synthesized a novel copolymer, poly(lactide)-D-α-tocopheryl polyethylene glycol 1000 succinate (PLA-TPGS), and found that it exhibited great potential in drug delivery with improved properties. In this study, we applied the Shirasu porous glass (SPG) membrane-emulsification technique to prepare the DTX-loaded PLA-TPGS nanoparticles on a pilot scale. The effect of several formulation variables on the DTX-loaded nanoparticle properties, including particle size, zeta potential, and drug-encapsulation efficiency, were investigated based on surfactant type and concentration in the aqueous phase, organic/aqueous phase volumetric ratio, membrane-pore size, transmembrane cycles, and operation pressure. The DTX-loaded nanoparticles were obtained with sizes of 306.8 ± 5.5 nm and 334.1 ± 2.7 nm (mean value ± standard deviation), and drug-encapsulation efficiency of 81.8% ± 4.5% and 64.5% ± 2.7% for PLA-TPGS and poly(lactic-co-glycolic acid) (PLGA) nanoparticles, respectively. In vivo pharmacokinetic study exhibited a significant advantage of PLA-TPGS nanoparticles over PLGA nanoparticles and Taxotere. Drug-loaded PLA-TPGS nanoparticles exhibited 1.78-, 6.34- and 3.35-fold higher values for area under the curve, half-life, and mean residence time, respectively, compared with those of PLGA nanoparticles, and 2.23-, 13.2-, 8.51-fold higher than those of Taxotere, respectively. In vivo real-time distribution of nanoparticles was measured on tumor-bearing mice by near-infrared fluorescence imaging, which demonstrated that the PLA-TPGS nanoparticles achieved much higher concentration and longer retention in tumors than PLGA nanoparticles after intravenous injection. This is consistent with the pharmacokinetic behavior of the nanoparticles. The tumor-inhibitory effect of DTX-loaded nanoparticles was observed in vivo in an H22 tumor-bearing mice model via intravenous administration. This indicated that PLA-TPGS nanoparticles are a feasible drug-delivery formulation with a pilot fabrication technique and have superior pharmacokinetic and anticancer effects compared to the commercially available Taxotere.
Collapse
Affiliation(s)
- Yunni Yu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
2017
|
Orozco VH, Palacio J, Sierra J, López BL. Increased covalent conjugation of a model antigen to poly(lactic acid)-g-maleic anhydride nanoparticles compared to bare poly(lactic acid) nanoparticles. Colloid Polym Sci 2013. [DOI: 10.1007/s00396-013-3023-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
2018
|
Whiting BT, Coates GW. Synthesis and Polymerization of Bicyclic Ketals: A Practical Route to High-Molecular Weight Polyketals. J Am Chem Soc 2013; 135:10974-7. [DOI: 10.1021/ja405581r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Bryan T. Whiting
- Department of Chemistry
and Chemical Biology, Baker
Laboratory, Cornell University, Ithaca,
New York 14853-1301, United States
| | - Geoffrey W. Coates
- Department of Chemistry
and Chemical Biology, Baker
Laboratory, Cornell University, Ithaca,
New York 14853-1301, United States
| |
Collapse
|
2019
|
Koenig O, Walker T, Perle N, Zech A, Neumann B, Schlensak C, Wendel HP, Nolte A. New aspects of gene-silencing for the treatment of cardiovascular diseases. Pharmaceuticals (Basel) 2013; 6:881-914. [PMID: 24276320 PMCID: PMC3816708 DOI: 10.3390/ph6070881] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 06/15/2013] [Accepted: 07/11/2013] [Indexed: 01/17/2023] Open
Abstract
Coronary heart disease (CHD), mainly caused by atherosclerosis, represents the single leading cause of death in industrialized countries. Besides the classical interventional therapies new applications for treatment of vascular wall pathologies are appearing on the horizon. RNA interference (RNAi) represents a novel therapeutic strategy due to sequence-specific gene-silencing through the use of small interfering RNA (siRNA). The modulation of gene expression by short RNAs provides a powerful tool to theoretically silence any disease-related or disease-promoting gene of interest. In this review we outline the RNAi mechanisms, the currently used delivery systems and their possible applications to the cardiovascular system. Especially, the optimization of the targeting and transfection procedures could enhance the efficiency of siRNA delivery drastically and might open the way to clinical applicability. The new findings of the last years may show the techniques to new innovative therapies and could probably play an important role in treating CHD in the future.
Collapse
Affiliation(s)
- Olivia Koenig
- Clinical Research Laboratory, Dept. of Thoracic, Cardiac and Vascular Surgery, University Hospital Tuebingen, Calwerstr. 7/1, 72076 Tuebingen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
2020
|
Elzoghby AO, Saad NI, Helmy MW, Samy WM, Elgindy NA. Ionically-crosslinked milk protein nanoparticles as flutamide carriers for effective anticancer activity in prostate cancer-bearing rats. Eur J Pharm Biopharm 2013; 85:444-51. [PMID: 23872177 DOI: 10.1016/j.ejpb.2013.07.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 06/14/2013] [Accepted: 07/09/2013] [Indexed: 02/07/2023]
Abstract
In this study, casein (CAS) nanoparticles were used to encapsulate the hydrophobic anticancer drug, flutamide (FLT), aiming at controlling its release, enhancing its anti-tumor activity, and reducing its hepatotoxicity. The nanoparticles were prepared by emulsification of CAS, at pH below its isoelectric point, and stabilized via ionic-crosslinking with sodium tripolyphosphate (TPP). The nanoparticles were spherical and positively charged with a size below 100 nm and exhibited a sustained drug release up to 4 days. After intravenous administration into prostate cancer-bearing rats for 28 days, FLT-loaded CAS nanoparticles showed a higher anti-tumor efficacy as revealed by a significantly higher % reduction in PSA serum level (75%) compared to free FLT (55%). Moreover, the nanoparticles demonstrated a marked reduction in the relative weights of both prostate tumor and seminal vesicle (43% and 32%) compared to free FLT (12% and 18%), respectively. A significantly higher anti-proliferative, anti-angiogenic, and apoptotic effects was demonstrated by the nanoparticles compared to drug solution as evidenced by their ability to decrease the expression of the proliferative marker (Ki-67) and reduce the level of tumor angiogenic markers (VEGF and IGF-1) as well as their ability to activate caspase-3 with subsequent induction of apoptosis in prostate cancer cells. Conclusively, these novel ionically-crosslinked milk protein nanovehicles offer a promising carrier to allow controlled intravenous delivery of hydrophobic anticancer drugs.
Collapse
Affiliation(s)
- Ahmed O Elzoghby
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | | | | | | | | |
Collapse
|
2021
|
Singh J, Chhabra G, Pathak K. Development of acetazolamide-loaded, pH-triggered polymeric nanoparticulatein situgel for sustained ocular delivery:in vitro. ex vivoevaluation and pharmacodynamic study. Drug Dev Ind Pharm 2013; 40:1223-32. [DOI: 10.3109/03639045.2013.814061] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
2022
|
Chen YC, Lo CL, Hsiue GH. Multifunctional nanomicellar systems for delivering anticancer drugs. J Biomed Mater Res A 2013; 102:2024-38. [PMID: 23828850 DOI: 10.1002/jbm.a.34850] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 06/10/2013] [Indexed: 12/26/2022]
Abstract
Most anticancer drugs cause severe side effect due to the lack of selectivity for cancer cells. In recent years, new strategies of micellar systems, which design for specifically target anticancer drugs to tumors, are developed at the forefront of polymeric science. To improve efficiency of delivery and cancer specificity, considerable emphasis has been placed on the development of micellar systems with passive and active targeting. In this review article, we summarized various strategies of designing multifunctional micellar systems in the purpose of improving delivery efficiency. Micellar systems compose of a multifunctional copolymer or a mixture of two or more copolymers with different properties is a plausible approach to tuning the resulting properties and satisfied various requirements for anticancer drug delivery. It appears that multifunctional micellar systems hold great potential in cancer therapy.
Collapse
Affiliation(s)
- Yi-Chun Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan, ROC; Department of Chemical Engineering and R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, 320, Taiwan, ROC
| | | | | |
Collapse
|
2023
|
Shilpa J, Anitha M, Paulose CS. Increased neuronal survival in the brainstem during liver injury: role of γ-aminobutyric acid and serotonin chitosan nanoparticles. J Neurosci Res 2013; 91:1203-14. [PMID: 23861071 DOI: 10.1002/jnr.23243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 03/12/2013] [Accepted: 03/27/2013] [Indexed: 11/07/2022]
Abstract
γ-Aminobutyric acid (GABA)- and serotonin (5-HT)-mediated cell signaling, neuronal survival enhancement, and reduced neuronal death in brainstem during liver injury followed by active liver regeneration have a critical role in maintaining routine bodily functions. In the present study, GABAB and 5-HT2A receptor functional regulation, interrelated actions of neuronal survival factors, and expression of apoptotic factors in the brainstem during GABA and 5-HT chitosan nanoparticles-induced active liver regeneration in partially hepatectomized rats were evaluated. Partially hepatectomized rats were treated with the nanoparticles, and receptor assays and confocal microscopic studies of GABAB and 5-HT2A receptors, gene expression studies of GABAB and 5-HT2A receptors, nuclear factor-κB (NF-κB), tumor necrosis factor-α (TNF-α), Akt-1, phospholipase C, Bax, and caspase-8 were performed with the brainstems of experimental animals. A significant decrease in GABAB and 5-HT2A receptor numbers and gene expressions denoted a homeostatic adjustment by the brain to trigger the sympathetic innervations during elevated DNA synthesis in the liver. The neuronal apoptosis resulting from the loss of liver function after partial hepatectomy was minimized by nanoparticle treatment in rats compared with rats with no treatment during regeneration. This was confirmed from the gene expression patterns of NF-κB, TNF-α, Akt-1, phospholipase C, Bax, and caspase-8. The present study revealed the potential of GABA and 5-HT chitosan nanoparticles for increasing neuronal survival in the brainstem during liver injury following regeneration, which avoids many neuropsychiatric problems.
Collapse
Affiliation(s)
- J Shilpa
- Molecular Neurobiology and Cell Biology Unit, Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology, Cochin, Kerala, India
| | | | | |
Collapse
|
2024
|
Guo M, Rong WT, Hou J, Wang DF, Lu Y, Wang Y, Yu SQ, Xu Q. Mechanisms of chitosan-coated poly(lactic-co-glycolic acid) nanoparticles for improving oral absorption of 7-ethyl-10-hydroxycamptothecin. NANOTECHNOLOGY 2013; 24:245101. [PMID: 23702815 DOI: 10.1088/0957-4484/24/24/245101] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Chitosan-modified poly(lactic-co-glycolic acid) nanoparticles (CHI/PLGA NPs) loaded with 7-ethyl-10-hydroxycamptothecin (SN-38), named CHI/PLGA/SN-38 NPs, were successfully prepared using an oil-in-water (O/W) solvent evaporation method. The physicochemical properties of the novel NPs were characterized by DLS, Zeta potential, SEM, DSC, XRD, and FTIR. The encapsulation efficiency and drug loading content were 71.83 (±2.77)% and 6.79 (±0.26)%, respectively. In vitro drug release in the simulated gastric juice was lower than that in the intestinal juice. In situ single-pass intestinal perfusion (SPIP) studies indicated a dramatic improvement of drug absorption as a result of the synergistic effect between CHI and PLGA on P-glycoprotein (Pgp) inhibition. CHI/PLGA NPs showed high cellular uptake and low efflux for drugs in Caco-2 cells. The cytotoxicity studies revealed that CHI/PLGA NPs had a transient effect on the membrane integrity, but did not have an influence on cell viability. Based on the in vitro release studies, SPIP, and intracellular drug accumulation and transport investigations, we speculate rationally that CHI/PLGA NPs were mainly internalized in the form of intact NPs, thus escaping the recognition of enterocyte Pgp and avoiding efflux into the apical part of the enterocytes. After partial release of drugs inside the enterocytes, CHI/PLGA interfered with the microenvironment of Pgp and further weakened the Pgp-mediated efflux. Then, the drug-loaded NPs exited via the exocytose effect from the basal part of the enterocytes and entered the blood circulation. These results showed that CHI/PLGA NPs would be smart oral delivery carriers for antineoplastic agents that are also Pgp substrates.
Collapse
Affiliation(s)
- Miao Guo
- Jiangsu Key Laboratory for Supramolecular Medicinal Materials and Applications, College of Life Sciences, Nanjing Normal University, Nanjing 210023, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
2025
|
Mody KT, Popat A, Mahony D, Cavallaro AS, Yu C, Mitter N. Mesoporous silica nanoparticles as antigen carriers and adjuvants for vaccine delivery. NANOSCALE 2013; 5:5167-79. [PMID: 23657437 DOI: 10.1039/c3nr00357d] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Vaccines have been at the forefront of improving human health for over two centuries. The challenges faced in developing effective vaccines flow from complexities associated with the immune system and requirement of an efficient and safe adjuvant to induce a strong adaptive immune response. Development of an efficient vaccine formulation requires careful selection of a potent antigen, efficient adjuvant and route of delivery. Adjuvants are immunological agents that activate the antigen presenting cells (APCs) and elicit a strong immune response. In the past decade, the use of mesoporous silica nanoparticles (MSNs) has gained significant attention as potential delivery vehicles for various biomolecules. In this review, we aim to highlight the potential of MSNs as vaccine delivery vehicles and their ability to act as adjuvants. We have provided an overview on the latest progress on synthesis, adsorption and release kinetics and biocompatibility of MSNs as next generation antigen carriers and adjuvants. A comprehensive summary on the ability of MSNs to deliver antigens and elicit both humoral and cellular immune responses is provided. Finally, we give insight on fundamental challenges and some future prospects of these nanoparticles as adjuvants.
Collapse
Affiliation(s)
- Karishma T Mody
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Australia
| | | | | | | | | | | |
Collapse
|
2026
|
Rescignano N, Tarpani L, Tiribuzi R, Montesano S, Martino S, Latterini L, Kenny JM, Armentano I. Protein encapsulation in biodegradable polymeric nanoparticles: morphology, fluorescence behaviour and stem cell uptake. Macromol Biosci 2013; 13:1204-12. [PMID: 23776101 DOI: 10.1002/mabi.201300140] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 04/03/2013] [Indexed: 11/12/2022]
Abstract
The synthesis and characterization of new biodegradable polymeric NPs loaded with bovine serum albumin marked with fluorescein isothiocyanate (FITC-BSA) is reported. The protein is encapsulated in poly(D,L-lactide-co-glycolide) (PLGA) NPs by the double emulsion method with subsequent solvent evaporation. The NPs display a spherical shape with a narrow size distribution and no aggregation is observed after drying. Steady-state and time-resolved fluorescence measurements appear to be a sensitive method to investigate the protein environment on the nanometer-scale. Finally, FITC-BSA-loaded NPs are rapidly internalized in stem cells. Interestingly, 25% cells were slightly positive after 28 days.
Collapse
Affiliation(s)
- Nicoletta Rescignano
- Materials Engineering Center, UdR INSTM, University of Perugia, Str. Pentima 4, 05100, Terni, Italy; Institute of Polymer Science and Technology, CSIC, Juan de la Cierva 3, 28006, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
2027
|
Novel Topical Ophthalmic Formulations for Management of Glaucoma. Pharm Res 2013; 30:2818-31. [DOI: 10.1007/s11095-013-1109-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 06/04/2013] [Indexed: 10/26/2022]
|
2028
|
Scalable method to produce biodegradable nanoparticles that rapidly penetrate human mucus. J Control Release 2013; 170:279-86. [PMID: 23751567 DOI: 10.1016/j.jconrel.2013.05.035] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/29/2013] [Accepted: 05/30/2013] [Indexed: 11/22/2022]
Abstract
Mucus typically traps and rapidly removes foreign particles from the airways, gastrointestinal tract, nasopharynx, female reproductive tract and the surface of the eye. Nanoparticles capable of rapid penetration through mucus can potentially avoid rapid clearance, and open significant opportunities for controlled drug delivery at mucosal surfaces. Here, we report an industrially scalable emulsification method to produce biodegradable mucus-penetrating particles (MPP). The emulsification of diblock copolymers of poly(lactic-co-glycolic acid) and polyethylene glycol (PLGA-PEG) using low molecular weight (MW) emulsifiers forms dense brush PEG coatings on nanoparticles that allow rapid nanoparticle penetration through fresh undiluted human mucus. In comparison, conventional high MW emulsifiers, such as polyvinyl alcohol (PVA), interrupts the PEG coating on nanoparticles, resulting in their immobilization in mucus owing to adhesive interactions with mucus mesh elements. PLGA-PEG nanoparticles with a wide range of PEG MW (1, 2, 5, and 10 kDa), prepared by the emulsification method using low MW emulsifiers, all rapidly penetrated mucus. A range of drugs, from hydrophobic small molecules to hydrophilic large biologics, can be efficiently loaded into biodegradable MPP using the method described. This readily scalable method should facilitate the production of MPP products for mucosal drug delivery, as well as potentially longer-circulating particles following intravenous administration.
Collapse
|
2029
|
Patel PJ, Acharya NS, Acharya SR. Development and characterization of glutathione-conjugated albumin nanoparticles for improved brain delivery of hydrophilic fluorescent marker. Drug Deliv 2013; 20:143-55. [DOI: 10.3109/10717544.2013.801050] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
2030
|
Patil GB, Surana SJ. Fabrication and statistical optimization of surface engineered PLGA nanoparticles for naso-brain delivery of ropinirole hydrochloride: in-vitro-ex-vivo studies. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2013; 24:1740-56. [PMID: 23705812 DOI: 10.1080/09205063.2013.798880] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Ropinirole hydrochloride (RPN), a nonergot dopamine D2-agonist used in the management of Parkinson's disease, has poor oral bioavailability (52%) due to extensive hepatic metabolism. The intent of present research work was aimed at design and statistical optimization of RPN-loaded poly (lactic-co-glycolic acid) (PLGA)-based biodegradable nanoparticles (NPs) surface modified using natural emulsifier, vitamin E (d-α-tocopheryl polyethylene glycol 1000 succinate [TPGS]) for direct nose-to-brain delivery in order to avoid hepatic first-pass metabolism, and improve therapeutic efficacy with sustained drug release. RPN-NPs were prepared by modified nanoprecipitation technique and optimized using 2(3) factorial design of experiment. The effect of polymer and emulsifier concentration was evaluated on particle size and entrapment efficiency (EE%). Formulation PL6 was considered as desirable with highest EE% (72.3 ± 6.1%), PS (279.4 ± 1.8 nm), zeta potential (-29.4 ± 2.6 mV), and cumulative drug diffusion of 96.43 ± 3.1% in 24 h. The ANOVA results for the dependent variables demonstrated that the model was significant (p value < 0.05) for response variables. Histopathological study of optimized batch (PL6) demonstrated good retention of NPs with no severe signs of damage on the integrity of nasal mucosa. Differential scanning calorimetry revealed the absence of any chemical interaction between RPN, PLGA, and TPGS while SEM study confirmed spherical shape of optimized NPs. Accelerated stability studies of freeze-dried optimized batch demonstrated negligible change in the average PS and EE% after storage at 25 ± 2 °C/60 ± 5% (relative humidity (RH) for the period of three months. The promising results of optimized batch suggested practicability of investigated system for enhancement of bioavailability and brain targeting of CNS acting drugs like RPN.
Collapse
Affiliation(s)
- Ganesh B Patil
- a Department of Pharmaceutics , R.C. Patel Institute of Pharmaceutical Education and Research , Shirpur, Dhule , India
| | | |
Collapse
|
2031
|
Khodir WKWA, Guarino V, Alvarez-Perez MA, Cafiero C, Ambrosio L. Trapping tetracycline-loaded nanoparticles into polycaprolactone fiber networks for periodontal regeneration therapy. J BIOACT COMPAT POL 2013. [DOI: 10.1177/0883911513481133] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The controlled delivery of antibiotics, anti-inflammatory agents, or chemotherapeutic agents to the periodontal site is a recognized strategy to improve the efficiency of regenerative processes of hard tissues. A novel approach based on the trapping of tetracycline hydrochloride–loaded particles in polycaprolactone nanofibers was used to guide the regeneration processes of periodontal tissue at the gum interface. Chitosan nanoparticles loaded with different levels of tetracycline hydrochloride (up to 5% wt) were prepared by solution nebulization induced by electrical forces (i.e. electrospraying). The fine tuning of process parameters allows to obtain nanoparticles with tailored sizes ranging from 0.485 ± 0.147 µm to 0.639 ± 0.154 µm. The tetracycline hydrochloride release profile had a predominant burst effect for the first 70% of release followed by a relatively slow release over 24 h, which is promising for oral drug delivery. We also demonstrated that trapping tetracycline hydrochloride–loaded particles with submicrometer diameters into a polycaprolactone fiber network contributed to slowing the release of tetracycline hydrochloride from the nanoparticles, thus providing a more prolonged release in the periodontal pocket during clinical therapy. Preliminary studies on human mesenchymal stem cells confirm the viability of cells up to 5 days after culture, and thereby, validate the use of nanoparticle-/nanofiber-integrated systems in periodontal therapies.
Collapse
Affiliation(s)
- WK Wan Abdul Khodir
- Institute of Composite and Biomedical Materials, National Research Council of Italy, Naples, Italy
| | - V Guarino
- Institute of Composite and Biomedical Materials, National Research Council of Italy, Naples, Italy
| | - MA Alvarez-Perez
- Institute of Composite and Biomedical Materials, National Research Council of Italy, Naples, Italy
| | - C Cafiero
- Department of Dentistry and Maxillo/Facial Surgery, Naples, Italy
| | - L Ambrosio
- Institute of Composite and Biomedical Materials, National Research Council of Italy, Naples, Italy
| |
Collapse
|
2032
|
Ma J, DU LF, Chen M, Wang HH, Xing LX, Jing LF, Li YH. Drug-loaded nano-microcapsules delivery system mediated by ultrasound-targeted microbubble destruction: A promising therapy method. Biomed Rep 2013; 1:506-510. [PMID: 24648976 DOI: 10.3892/br.2013.110] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 04/15/2013] [Indexed: 11/06/2022] Open
Abstract
The nano-microcapsules drug delivery system is currently a promising method for the treatment of many types of diseases, particularly tumors. However, the drug delivery efficiency does not reach a satisfactory level to meet treatment demands. Therefore, the effectiveness of delivery needs to be improved. Based on the alterations in the structure and modification of nano-microcapsules, ultrasound-targeted microbubble destruction (UTMD), a safe physical targeted method, may increase tissue penetration and cell membrane permeability, aiding the drug-loaded nano-microcapsules ingress the interior of targeted tissues and cells. The effectiveness and exact mechanism of action of the drug-loaded nano-microcapsules delivery system mediated by UTMD have yet to be fully elucidated. In this study, the latest advancement in UTMD-mediated drug loaded nano-microcapsules system technology was reviewed and the hindrances of UTMD-mediated drug delivery were assessed, in combination with a prospective study. The findings suggested that the drug delivery efficiency of nano-microcapsules mediated by UTMD was distinctly improved. Thus, the UTMD-mediated drug-loaded nano-microcapsules delivery system may significantly improve the efficiency of drug delivery, which may be a promising new therapeutic method.
Collapse
Affiliation(s)
- Jing Ma
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080; ; Department of Cardiovascular Ultrasound, Shanghai East Hospital Affiliated to Tongji University, Shanghai 200120, P.R. China
| | - Lian Fang DU
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080
| | - Ming Chen
- Department of Cardiovascular Ultrasound, Shanghai East Hospital Affiliated to Tongji University, Shanghai 200120, P.R. China
| | - Hang Hui Wang
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080
| | - Ling Xi Xing
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080
| | - Li Fang Jing
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080
| | - Yun Hua Li
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080
| |
Collapse
|
2033
|
Elzoghby AO, Helmy MW, Samy WM, Elgindy NA. Novel ionically crosslinked casein nanoparticles for flutamide delivery: formulation, characterization, and in vivo pharmacokinetics. Int J Nanomedicine 2013; 8:1721-32. [PMID: 23658490 PMCID: PMC3647443 DOI: 10.2147/ijn.s40674] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A novel particulate delivery matrix based on ionically crosslinked casein (CAS) nanoparticles was developed for controlled release of the poorly soluble anticancer drug flutamide (FLT). Nanoparticles were fabricated via oil-in-water emulsification then stabilized by ionic crosslinking of the positively charged CAS molecules below their isoelectric point, with the polyanionic crosslinker sodium tripolyphosphate. With the optimal preparation conditions, the drug loading and incorporation efficiency achieved were 8.73% and 64.55%, respectively. The nanoparticles exhibited a spherical shape with a size below 100 nm and a positive zeta potential (+7.54 to +17.3 mV). FLT was molecularly dispersed inside the nanoparticle protein matrix, as revealed by thermal analysis. The biodegradability of CAS nanoparticles in trypsin solution could be easily modulated by varying the sodium tripolyphosphate crosslinking density. A sustained release of FLT from CAS nanoparticles for up to 4 days was observed, depending on the crosslinking density. After intravenous administration of FLT-CAS nanoparticles into rats, CAS nanoparticles exhibited a longer circulation time and a markedly delayed blood clearance of FLT, with the half-life of FLT extended from 0.88 hours to 14.64 hours, compared with drug cosolvent. The results offer a promising method for tailoring biodegradable, drug-loaded CAS nanoparticles as controlled, long-circulating drug delivery systems of hydrophobic anticancer drugs in aqueous vehicles.
Collapse
Affiliation(s)
- Ahmed O Elzoghby
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | | | | | | |
Collapse
|
2034
|
Bulcão RP, de Freitas FA, Dallegrave E, Venturini CG, Baierle M, Durgante J, Sauer E, Cassini C, Cerski CT, Zielinsky P, Salvador M, Pohlmann AR, Guterres SS, Garcia SC. In vivo toxicological evaluation of polymeric nanocapsules after intradermal administration. Eur J Pharm Biopharm 2013; 86:167-77. [PMID: 23643792 DOI: 10.1016/j.ejpb.2013.04.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 03/14/2013] [Accepted: 04/05/2013] [Indexed: 12/11/2022]
Abstract
Polymeric nanocarriers have shown great promise as delivery systems. An alternative strategy has been to explore new delivery routes, such as intradermal (i.d.), that can be used for vaccines and patch-based drug delivery. Despite their many advantages, there are few toxicity studies, especially in vivo. We report a safety assessment of biodegradable poly(ɛ-caprolactone) lipid-core nanocapsules (LNC) with a mean size of 245±10nm following single and repeated intradermal injections to Wistar rats. Suspensions were prepared by interfacial deposition of polymer. The animals (n=6/group) received a single-dose of saline solution (1.2ml/kg) or LNC (7.2×10(12)LNC/kg), or repeated-doses of two controls, saline solution or Tween 80 (0.9ml/kg), or three different concentrations of LNC (1.8, 3.6, and 5.4×10(12)LNC/kg) for 28 consecutive days. Clinical and physiological signs and mortality were observed. Samples of urine, blood, and tissue were used to perform toxicological evaluation. There were no clinical signs of toxicity or mortality, but there was a slight decrease in the relative body weights in the Tween 80-treated group (p<0.01) after repeated administration. No histopathological alterations were observed in tissues or significant changes in blood and urinary biomarkers for tissue damage. Mild alterations in white blood cells count with increases in granulocytes in the Tween-80 group (p<0.05) were found. Genotoxicity was evaluated through the comet assay, and no statistical difference was observed among the groups. Therefore, we conclude that, under the conditions of these experiments, biodegradable LNC did not present appreciable toxicity after 28 consecutive days of intradermal administration and is promising for its future application in vaccines and patch-based devices for enhancing the delivery of drugs.
Collapse
Affiliation(s)
- Rachel P Bulcão
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Brazil; Laboratório de Toxicologia (LATOX), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernando A de Freitas
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Brazil; Laboratório de Toxicologia (LATOX), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eliane Dallegrave
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Cristina G Venturini
- Departamento de Produção e Controle de Medicamentos, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marília Baierle
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Brazil; Laboratório de Toxicologia (LATOX), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Juliano Durgante
- Laboratório de Toxicologia (LATOX), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Elisa Sauer
- Laboratório de Toxicologia (LATOX), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Instituto de Cardiologia, Fundação Universitária de Cardiologia, Porto Alegre, Brazil
| | - Carina Cassini
- Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Brazil
| | - Carlos T Cerski
- Departamento de Patologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Paulo Zielinsky
- Instituto de Cardiologia, Fundação Universitária de Cardiologia, Porto Alegre, Brazil
| | - Mirian Salvador
- Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Brazil
| | - Adriana R Pohlmann
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Brazil; Departamento de Química Orgânica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sílvia S Guterres
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Brazil; Departamento de Produção e Controle de Medicamentos, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Solange C Garcia
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Brazil; Laboratório de Toxicologia (LATOX), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
2035
|
Xiang SD, Wilson K, Day S, Fuchsberger M, Plebanski M. Methods of effective conjugation of antigens to nanoparticles as non-inflammatory vaccine carriers. Methods 2013; 60:232-41. [PMID: 23643867 DOI: 10.1016/j.ymeth.2013.03.036] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/24/2013] [Accepted: 03/27/2013] [Indexed: 11/25/2022] Open
Abstract
It has recently become clear that nanoparticle size is a major determinant for how antigen presenting cells (APCs), and specifically dendritic cells (DC) recognize and handle particles, and hence a critical parameter for the formulation of particulate vaccines that aim to induce immunity by targeting DC. Our previous studies in mice and sheep have shown polystyrene nanoparticles of 40-50 nm (PSNPs) with covalently bound antigen offer a new class of vaccines, which contain only 2 elements, antigen and particle, and no added inflammatory stimuli, but evoke very potent combined CD8 T cell and antibody responses. Herein we have optimized the methods for antigen conjugation to PSNPs to controllably promote a single antigen (protein or peptide) layer coating on the nanoparticle. Surprisingly, these nanovaccines not only continued to induce high levels of CD8 T cells in vivo, but were further more potent antibody inducers than nanoparticles containing multiple antigen layers. Addressing the issue of antigen loading on PSNPs, we found an optimal range, above or below which immunogenicity is changed either for antibodies or CD8 T cells. The mechanism behind the induction of high levels of CD8 T cells was further explored by assessing the DC subset that takes up the PSNPs in vivo, and these were found to be preferentially CD8(+) CD11c(+) DC in the lymph node draining the injection site. Since the levels of induced antibodies were highly elevated, and CD8(+) DC do not traditionally induce antibodies, we further sought to find if, despite no detectable inflammation at the injection site, the PSNPs may perhaps induce inflammatory cytokines locally in the lymph node after injection, or systemically in sera, resulting in an adjuvant effect. The initial findings presented herein show no detectable induction of the key inflammatory cytokines such as TNF-α, IL-1 or IL-6, suggesting a novel "non-inflammatory" adjuvant mechanism.
Collapse
Affiliation(s)
- Sue D Xiang
- Department of Immunology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, 89 Commercial Rd., Melbourne, Victoria 3004, Australia.
| | | | | | | | | |
Collapse
|
2036
|
Aguzzi C, Viseras C, Cerezo P, Salcedo I, Sánchez-Espejo R, Valenzuela C. Release kinetics of 5-aminosalicylic acid from halloysite. Colloids Surf B Biointerfaces 2013; 105:75-80. [DOI: 10.1016/j.colsurfb.2012.12.041] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 12/16/2012] [Indexed: 12/01/2022]
|
2037
|
Giri TK, Thakur D, Alexander A, Badwaik H, Tripathy M, Tripathi DK. Biodegradable IPN hydrogel beads of pectin and grafted alginate for controlled delivery of diclofenac sodium. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2013; 24:1179-1190. [PMID: 23423649 DOI: 10.1007/s10856-013-4884-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 02/04/2013] [Indexed: 06/01/2023]
Abstract
A novel diclofenac sodium (DS) loaded interpenetrating polymer network (IPN) beads of pectin and hydrolyzed polyacrylamide-graft-sodium alginate (PAAm-g-SA) was developed through ionotropic gelation and covalent cross-linking. The graft copolymer was synthesized by free radical polymerization under the nitrogen atmosphere followed by alkaline hydrolysis. The grafting, alkaline hydrolysis, and characterization of beads were confirmed by Fourier transforms infrared spectroscopy. The crystalline structure of drug after encapsulation into IPN beads were evaluated by differential scanning colorimetry and X-ray diffraction analyses. DS encapsulation was up to 96.45 %. The effect of hydrolyzed graft copolymer/pectin ratios and glutaraldehyde concentration on drug release in acidic and phosphate buffer solutions were investigated. The release of drug was significantly increased with increase of pH. The release of drug depends on the extent of cross-linking. The results indicated that IPN beads of hydrolyzed PAAm-g-SA and pectin could be used for sustained release of DS.
Collapse
Affiliation(s)
- Tapan Kumar Giri
- Rungta College of Pharmaceutical Sciences and Research, Kohka Road, Kurud, Bhilai 491024, India.
| | | | | | | | | | | |
Collapse
|
2038
|
Manish M, Rahi A, Kaur M, Bhatnagar R, Singh S. A single-dose PLGA encapsulated protective antigen domain 4 nanoformulation protects mice against Bacillus anthracis spore challenge. PLoS One 2013; 8:e61885. [PMID: 23637922 PMCID: PMC3639271 DOI: 10.1371/journal.pone.0061885] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 03/14/2013] [Indexed: 12/11/2022] Open
Abstract
Bacillus anthracis, the etiological agent of anthrax, is a major bioterror agent. Vaccination is the most effective prophylactic measure available against anthrax. Currently available anthrax vaccines have issues of the multiple booster dose requirement, adjuvant-associated side effects and stability. Use of biocompatible and biodegradable nanoparticles to deliver the antigens to immune cells could solve the issues associated with anthrax vaccines. We hypothesized that the delivery of a stable immunogenic domain 4 of protective antigen (PAD4) of Bacillus anthracis encapsulated in a poly (lactide-co-glycolide) (PLGA)--an FDA approved biocompatible and biodegradable material, may alleviate the problems of booster dose, adjuvant toxicity and stability associated with anthrax vaccines. We made a PLGA based protective antigen domain 4 nanoparticle (PAD4-NP) formulation using water/oil/water solvent evaporation method. Nanoparticles were characterized for antigen content, morphology, size, polydispersity and zeta potential. The immune correlates and protective efficacy of the nanoparticle formulation was evaluated in Swiss Webster outbred mice. Mice were immunized with single dose of PAD4-NP or recombinant PAD4. The PAD4-NP elicited a robust IgG response with mixed IgG1 and IgG2a subtypes, whereas the control PAD4 immunized mice elicited low IgG response with predominant IgG1 subtype. The PAD4-NP generated mixed Th1/Th2 response, whereas PAD4 elicited predominantly Th2 response. When we compared the efficacy of this single-dose vaccine nanoformulation PAD4-NP with that of the recombinant PAD4 in providing protective immunity against a lethal challenge with Bacillus anthracis spores, the median survival of PAD4-NP immunized mice was 6 days as compared to 1 day for PAD4 immunized mice (p<0.001). Thus, we demonstrate, for the first time, the possibility of the development of a single-dose and adjuvant-free protective antigen based anthrax vaccine in the form of PAD4-NP. Further work in this direction may produce a better and safer candidate anthrax vaccine.
Collapse
Affiliation(s)
- Manish Manish
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Amit Rahi
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Manpreet Kaur
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Rakesh Bhatnagar
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Samer Singh
- Special Centre for Nano Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
2039
|
Zhang Z, Qu Q, Li J, Zhou S. The Effect of the Hydrophilic/Hydrophobic Ratio of Polymeric Micelles on their Endocytosis Pathways into Cells. Macromol Biosci 2013; 13:789-98. [DOI: 10.1002/mabi.201300037] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Indexed: 02/01/2023]
|
2040
|
Kim AJ, Boylan NJ, Suk JS, Hwangbo M, Yu T, Schuster BS, Cebotaru L, Lesniak WG, Oh JS, Adstamongkonkul P, Choi AY, Kannan RM, Hanes J. Use of single-site-functionalized PEG dendrons to prepare gene vectors that penetrate human mucus barriers. Angew Chem Int Ed Engl 2013; 52:3985-8. [PMID: 23460577 PMCID: PMC3782282 DOI: 10.1002/anie.201208556] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Indexed: 11/07/2022]
Affiliation(s)
- Anthony J. Kim
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University, Baltimore (USA)
- Department of Chemical & Biomolecular Engineering Johns Hopkins University, Baltimore (USA)
| | - Nicholas J. Boylan
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University, Baltimore (USA)
- Department of Chemical & Biomolecular Engineering Johns Hopkins University, Baltimore (USA)
| | - Jung Soo Suk
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University, Baltimore (USA)
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore (USA)
| | - Minyoung Hwangbo
- Department of Chemical & Biomolecular Engineering Johns Hopkins University, Baltimore (USA)
| | - Tao Yu
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University, Baltimore (USA)
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore (USA)
| | - Benjamin S. Schuster
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University, Baltimore (USA)
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore (USA)
| | - Liudimila Cebotaru
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University, Baltimore (USA)
| | - Wojciech G. Lesniak
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University, Baltimore (USA)
| | - Joon Seok Oh
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore (USA)
| | | | - Ashley Y. Choi
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University, Baltimore (USA)
| | - Rangaramanujam M. Kannan
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University, Baltimore (USA)
| | - Justin Hanes
- Departments of Ophthalmology, Biomedical Engineering, Chemical & Biomolecular Engineering and Oncology, Center for Cancer Nanotechnology Excellence, and Center for Nanomedicine, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231 (USA)
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University, Baltimore (USA)
- Department of Chemical & Biomolecular Engineering Johns Hopkins University, Baltimore (USA)
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore (USA)
| |
Collapse
|
2041
|
Ishak RA, Awad GA, Zaki NM, El-Shamy AEHA, Mortada ND. A comparative study of chitosan shielding effect on nano-carriers hydrophilicity and biodistribution. Carbohydr Polym 2013; 94:669-76. [DOI: 10.1016/j.carbpol.2013.01.072] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Revised: 12/04/2012] [Accepted: 01/23/2013] [Indexed: 01/28/2023]
|
2042
|
Della Porta G, Campardelli R, Reverchon E. Monodisperse biopolymer nanoparticles by Continuous Supercritical Emulsion Extraction. J Supercrit Fluids 2013. [DOI: 10.1016/j.supflu.2013.01.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
2043
|
Pinto AM, Moreira S, Gonçalves IC, Gama FM, Mendes AM, Magalhães FD. Biocompatibility of poly(lactic acid) with incorporated graphene-based materials. Colloids Surf B Biointerfaces 2013; 104:229-38. [DOI: 10.1016/j.colsurfb.2012.12.006] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 11/30/2012] [Accepted: 12/08/2012] [Indexed: 10/27/2022]
|
2044
|
|
2045
|
Zaric M, Lyubomska O, Touzelet O, Poux C, Al-Zahrani S, Fay F, Wallace L, Terhorst D, Malissen B, Henri S, Power UF, Scott CJ, Donnelly RF, Kissenpfennig A. Skin dendritic cell targeting via microneedle arrays laden with antigen-encapsulated poly-D,L-lactide-co-glycolide nanoparticles induces efficient antitumor and antiviral immune responses. ACS NANO 2013; 7:2042-55. [PMID: 23373658 PMCID: PMC3936823 DOI: 10.1021/nn304235j] [Citation(s) in RCA: 166] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 02/01/2013] [Indexed: 05/19/2023]
Abstract
The efficacious delivery of antigens to antigen-presenting cells (APCs), in particular, to dendritic cells (DCs), and their subsequent activation remains a significant challenge in the development of effective vaccines. This study highlights the potential of dissolving microneedle (MN) arrays laden with nanoencapsulated antigen to increase vaccine immunogenicity by targeting antigen specifically to contiguous DC networks within the skin. Following in situ uptake, skin-resident DCs were able to deliver antigen-encapsulated poly-d,l-lactide-co-glycolide (PGLA) nanoparticles to cutaneous draining lymph nodes where they subsequently induced significant expansion of antigen-specific T cells. Moreover, we show that antigen-encapsulated nanoparticle vaccination via microneedles generated robust antigen-specific cellular immune responses in mice. This approach provided complete protection in vivo against both the development of antigen-expressing B16 melanoma tumors and a murine model of para-influenza, through the activation of antigen-specific cytotoxic CD8(+) T cells that resulted in efficient clearance of tumors and virus, respectively. In addition, we show promising findings that nanoencapsulation facilitates antigen retention into skin layers and provides antigen stability in microneedles. Therefore, the use of biodegradable polymeric nanoparticles for selective targeting of antigen to skin DC subsets through dissolvable MNs provides a promising technology for improved vaccination efficacy, compliance, and coverage.
Collapse
Affiliation(s)
- Marija Zaric
- The Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, University Road, Belfast BT9 7AE, United Kingdom
| | - Oksana Lyubomska
- The Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, University Road, Belfast BT9 7AE, United Kingdom
| | - Olivier Touzelet
- The Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, University Road, Belfast BT9 7AE, United Kingdom
| | - Candice Poux
- The Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, University Road, Belfast BT9 7AE, United Kingdom
| | - Sharifah Al-Zahrani
- School of Pharmacy, Queen’s University Belfast, Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Francois Fay
- School of Pharmacy, Queen’s University Belfast, Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Leah Wallace
- The Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, University Road, Belfast BT9 7AE, United Kingdom
| | - Dorothea Terhorst
- Centre d’Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, UM2, Marseille, France
- INSERM U1104, Marseille, France
- CNRS UMR7280, Marseille, France
| | - Bernard Malissen
- Centre d’Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, UM2, Marseille, France
- INSERM U1104, Marseille, France
- CNRS UMR7280, Marseille, France
| | - Sandrine Henri
- Centre d’Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, UM2, Marseille, France
- INSERM U1104, Marseille, France
- CNRS UMR7280, Marseille, France
| | - Ultan F. Power
- The Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, University Road, Belfast BT9 7AE, United Kingdom
| | - Christopher J. Scott
- School of Pharmacy, Queen’s University Belfast, Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Ryan F. Donnelly
- School of Pharmacy, Queen’s University Belfast, Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Adrien Kissenpfennig
- The Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, University Road, Belfast BT9 7AE, United Kingdom
- Address correspondence to
| |
Collapse
|
2046
|
Kusonwiriyawong C, Lipipun V, Vardhanabhuti N, Zhang Q, Ritthidej GC. Spray-dried chitosan microparticles for cellular delivery of an antigenic protein: physico-chemical properties and cellular uptake by dendritic cells and macrophages. Pharm Res 2013; 30:1677-97. [PMID: 23483441 DOI: 10.1007/s11095-013-1014-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 02/15/2013] [Indexed: 11/29/2022]
Abstract
PURPOSE Spray-dried chitosan microparticles for cellular delivery of antigen to dendritic cells (DC) and macrophages (Mϕ) were investigated. METHODS Chitosan microparticles were prepared by spray drying. For comparison, poly(lactic-co-glycolic acid) (PLGA) and poly(α-butyl cyanoacrylate) (BCA) micro-/nanoparticles were generated. Bovine serum albumin (BSA) was used as a model antigen. The particles were characterized in terms of size, morphology, surface charge, surface composition, protein content, entrapment efficiency, in vitro release, and protein integrity. Additionally, they were subject to cell viability and cellular uptake study with DC and Mϕ. RESULTS Size of chitosan, PLGA, and BCA micro-/nanoparticles ranged between 3.11-7.18, 0.94-6.26, and 0.30-6.34 μm, respectively. Particle morphology and in vitro protein release varied, depending on polymer type, particle composition and preparation process parameters. Chitosan microparticles were cationic, while PLGA microparticles were neutral. BCA micro-/nanoparticles were either anionic or cationic, according to polymerization pH. Protein content and entrapment efficiency of chitosan and PLGA microparticles were relatively consistent. Only integrity and conformational structure of protein encapsulated in chitosan microparticles were completely retained. Chitosan and PLGA microparticles were non-toxic to DC and Mϕ, but the former were internalized more efficiently. CONCLUSIONS Spray-dried chitosan microparticles delivered the antigen efficiently to DC and Mϕ.
Collapse
Affiliation(s)
- Chirasak Kusonwiriyawong
- Department of Pharmaceutics and Industrial Pharmacy Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | | | | | | |
Collapse
|
2047
|
Vilos C, Morales FA, Solar PA, Herrera NS, Gonzalez-Nilo FD, Aguayo DA, Mendoza HL, Comer J, Bravo ML, Gonzalez PA, Kato S, Cuello MA, Alonso C, Bravo EJ, Bustamante EI, Owen GI, Velasquez LA. Paclitaxel-PHBV nanoparticles and their toxicity to endometrial and primary ovarian cancer cells. Biomaterials 2013; 34:4098-4108. [PMID: 23465827 DOI: 10.1016/j.biomaterials.2013.02.034] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 02/12/2013] [Indexed: 12/11/2022]
Abstract
This report is an integrated study to include the molecular simulation, physicochemical characterization and biological analysis of a paclitaxel-loaded PHBV nanoparticle that demonstrates uptake, release and cytotoxicity in cancer cell lines. Taking this nanoparticle one step closer to its use in a clinical setting, we demonstrate that it causes significant cell death in primary cultures of stage IIIc serous ovarian cancer cells isolated from six patients. Molecular simulations revealed a high affinity of paclitaxel for the water-polymer interface, thus the drug is delivered only when the polymer near it is degraded. The Fourier transform infrared spectroscopy suggests the formation of a short-lived crystalline phase, also observed in the CG simulations, and transmission electron microscopy revealed branched structures on the surface of particles, which disappeared after 4 days. Biological analyses indicated that these particles have a 48-h window of toxicity protection, allowing for the endocytosis of the particle by the cells; this finding was corroborated by confocal microscopy and flow cytometry. The low cost to synthesize PHBV using microorganisms and the potential chemical modifications of the polymer make it attractive for inexpensive, large-scale pharmaceutical production.
Collapse
Affiliation(s)
- Cristian Vilos
- Universidad Andres Bello, Facultad de Medicina, Center for Integrative Medicine and Innovative Science (CIMIS), Echaurren 183, Santiago, Chile; Centro para el Desarrollo de la Nanociencia y Nanotecnología, Universidad de Santiago de Chile, Ecuador 3493, Santiago, Chile
| | - Francisco A Morales
- Universidad Andres Bello, Facultad de Medicina, Center for Integrative Medicine and Innovative Science (CIMIS), Echaurren 183, Santiago, Chile
| | - Paula A Solar
- Universidad Andres Bello, Facultad de Medicina, Center for Integrative Medicine and Innovative Science (CIMIS), Echaurren 183, Santiago, Chile
| | - Natalia S Herrera
- Universidad Andres Bello, Facultad de Medicina, Center for Integrative Medicine and Innovative Science (CIMIS), Echaurren 183, Santiago, Chile
| | - Fernando D Gonzalez-Nilo
- Universidad Andres Bello, Facultad de Biología, Center for Bioinformatics and Integrative Biology (CBIB), Republica 239, Santiago, Chile
| | - Daniel A Aguayo
- Universidad Andres Bello, Facultad de Biología, Center for Bioinformatics and Integrative Biology (CBIB), Republica 239, Santiago, Chile
| | - Hegaly L Mendoza
- Universidad Andres Bello, Facultad de Biología, Center for Bioinformatics and Integrative Biology (CBIB), Republica 239, Santiago, Chile
| | - Jeffrey Comer
- Universidad Andres Bello, Facultad de Biología, Center for Bioinformatics and Integrative Biology (CBIB), Republica 239, Santiago, Chile; Fundación Fraunhofer Chile Research, M. Sánchez Fontecilla 310 piso 14, Las Condes, Chile
| | - Maria L Bravo
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | - Pamela A Gonzalez
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | - Sumie Kato
- Facultad de Medicina, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | - Mauricio A Cuello
- Facultad de Medicina, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | | | - Erasmo J Bravo
- Hospital Gustavo Fricke, Alvarez 1532, Viña del Mar, Chile
| | | | - Gareth I Owen
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | - Luis A Velasquez
- Universidad Andres Bello, Facultad de Medicina, Center for Integrative Medicine and Innovative Science (CIMIS), Echaurren 183, Santiago, Chile; Centro para el Desarrollo de la Nanociencia y Nanotecnología, Universidad de Santiago de Chile, Ecuador 3493, Santiago, Chile.
| |
Collapse
|
2048
|
Kim AJ, Boylan NJ, Suk JS, Hwangbo M, Yu T, Schuster BS, Cebotaru L, Lesniak WG, Oh JS, Adstamongkonkul P, Choi AY, Kannan RM, Hanes J. Use of Single-Site-Functionalized PEG Dendrons To Prepare Gene Vectors that Penetrate Human Mucus Barriers. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201208556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
2049
|
Wang Q, Bao Y, Ahire J, Chao Y. Co-encapsulation of biodegradable nanoparticles with silicon quantum dots and quercetin for monitored delivery. Adv Healthc Mater 2013. [PMID: 23184534 DOI: 10.1002/adhm.201200178] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Polymer nanoparticles have emerged as a promising new strategy for the efficient delivery of drugs. They have several advantages when used as drug carriers, such as high stability, high capacity, improvement of drug bioavailability, as well as allowing for sustained drug release. Quercetin has therapeutic potential as an anticancer drug, but has poor solubility and low bioavailability. In this study it is shown that co-encapsulation of quercetin and fluorescent Silicon quantum dots (SiQDs) in poly (ethylene glycol)-block-polylactide (PEG-PLA) nanoparticles can be used for simultaneous in vitro imaging and to improve the biocompatibility of quercetin. Fluorescent imaging with SiQDs can provide a new concept to monitor the delivery of anti-cancer drugs. The nanoparticles are synthesized based on the double emulsion method and are extensively characterized and assayed for cytotoxicity in vitro. HepG2 cells are incubated with quercetin and SiQDs dual-loaded PEG-PLA nanoparticles, resulting in a red fluorescent staining which can be detected with a confocal microscope. PEG-PLA nanoparticle encapsulated quercetin suppresses human hepatoma HepG2 cell proliferation more effectively than the free-standing form. In addition, nanoparticle-encapsulated quercetin significantly inhibits hydrogen peroxide-induced DNA damage in HepG2 cells. These data show that nanocapsulated quercetin possesses the potential bioactivity to reduce the drug dosage frequency, as well as increase patient compliance. The combination of polymeric nanoparticles and semiconductor quantum dots can allow monitoring of delivery, improve aqueous solubility, and enhance biocompatibility. Such nanoparticulated systems could shape the future of drug delivery.
Collapse
Affiliation(s)
- Qi Wang
- Energy Materials Laboratory, School of Chemistry, University of East Anglia, Norwich, NR4 7TJ, UK
| | | | | | | |
Collapse
|
2050
|
Effect of concentration and degree of saturation on co-precipitation of catechin and poly(l-lactide) by the RESOLV process. J Supercrit Fluids 2013. [DOI: 10.1016/j.supflu.2012.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|