301
|
Application of a Quality-By-Design Approach to Optimise Lipid-Polymer Hybrid Nanoparticles Loaded with a Splice-Correction Antisense Oligonucleotide: Maximising Loading and Intracellular Delivery. Pharm Res 2019; 36:37. [PMID: 30623253 DOI: 10.1007/s11095-018-2566-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 12/26/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Antisense oligonucleotides (ASOs) are promising therapeutics for specific modulation of cellular RNA function. However, ASO efficacy is compromised by inefficient intracellular delivery. Lipid-polymer hybrid nanoparticles (LPNs) are attractive mediators of intracellular ASO delivery due to favorable colloidal stability and sustained release properties. METHODS LPNs composed of cationic lipidoid 5 (L5) and poly(DL-lactic-co-glycolic acid) were studied for delivery of an ASO mediating splice correction of a luciferase gene transcript (Luc-ASO). Specific purposes were: (i) to increase the mechanistic understanding of factors determining the loading of ASO in LPNs, and (ii) to optimise the LPNs and customise them for Luc-ASO delivery in HeLa pLuc/705 cells containing an aberrant luciferase gene by using a quality-by-design approach. Critical formulation variables were linked to critical quality attributes (CQAs) using risk assessment and design of experiments, followed by delineation of an optimal operating space (OOS). RESULTS A series of CQAs were identified based on the quality target product profile. The L5 content and L5:Luc-ASO ratio (w/w) were determined as critical formulation variables, which were optimised systematically. The optimised Luc-ASO-loaded LPNs, defined from the OOS, displayed high loading and mediated splice correction at well-tolerated, lower doses as compared to those required for reference L5-based lipoplexes, L5-modified stable nucleic acid lipid nanoparticles or LPNs modified with dioleoyltrimethylammonium propane (conventional cationic lipid). CONCLUSIONS The optimal Luc-ASO-loaded LPNs represent a robust formulation that mediates efficient intracellular delivery of Luc-ASO. This opens new avenues for further development of LPNs as a broadly applicable technology platform for delivering nucleic acid cargos intracellularly.
Collapse
|
302
|
Nowalk JA, Swisher JH, Meyer TY. Consequences of isolated critical monomer sequence errors for the hydrolysis behaviors of sequenced degradable polyesters. Polym Chem 2019. [DOI: 10.1039/c9py00891h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Despite the known sensitivity to sequence mutations of biological polymers, little is known about the effects of errors in sequenced synthetic copolymers.
Collapse
Affiliation(s)
- Jamie A. Nowalk
- Department of Chemistry
- University of Pittsburgh
- Pittsburgh
- USA
| | | | - Tara Y. Meyer
- Department of Chemistry
- University of Pittsburgh
- Pittsburgh
- USA
- McGowan Institute for Regenerative Medicine
| |
Collapse
|
303
|
Gritsch L, Conoscenti G, La Carrubba V, Nooeaid P, Boccaccini AR. Polylactide-based materials science strategies to improve tissue-material interface without the use of growth factors or other biological molecules. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 94:1083-1101. [DOI: 10.1016/j.msec.2018.09.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 08/14/2018] [Accepted: 09/11/2018] [Indexed: 01/11/2023]
|
304
|
Han C, Li X, Zhou T, Chen C, Zhang K, Yang S, Wang X, Tian H, Zhao C, Zhao J. A tranilast and BMP-2 based functional bilayer membrane is effective for the prevention of epidural fibrosis during spinal lamina reconstruction. J Mater Chem B 2019. [DOI: 10.1039/c8tb03071e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Failed Back Surgery Syndrome (FBSS) is a common complication of lumbar surgery.
Collapse
|
305
|
Eskandari Z, Kazdal F, Bahadori F, Ebrahimi N. Quality-by-design model in optimization of PEG-PLGA nano micelles for targeted cancer therapy. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
306
|
Sun H, Liu CZ, Liu C, Tang M, Cao G, Zhang Q, Gu X. Employing the Sirolimus-Eluting Poly (Propylene Carbonate) Mesh for the Prevention of Arteriovenous Graft Stenosis in Rats. J Cardiovasc Pharmacol Ther 2018; 24:269-277. [PMID: 30474386 DOI: 10.1177/1074248418806060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Poly (propylene carbonate, PPC) is a new member of the aliphatic polyester family. An outstanding feature of PPC is that it produces mainly water and carbon dioxide when degraded in vivo, causing minimal side effects. This unique property together with excellent biocompatibility and biodegradability makes PPC a promising material for drug delivery. In this study, we explored the effect of the sirolimus (an inhibitor of cell growth)-eluting PPC mesh on graft stenosis and its possible mechanisms in a rat arteriovenous grafting model. The PPC mesh was prepared by electrospinning. A jugular vein to abdominal aortic autograft transplantation model was established in rats. The graft was then treated by wrapping with the drug mesh or the drug-free mesh or left untreated. Four weeks posttransplantation, neointima was measured with hematoxylin and eosin staining, matrix metalloproteinase-2 (MMP-2), and MMP-9, and proliferating cell nuclear antigen (PCNA) in the grafts were assayed by Western blotting and immunohistochemistry, respectively. In vitro rat aortic adventitial fibroblast cell (RAAFC) migration was assessed using the Boyden chamber assay, and phospho-mammalian target of rapamycin (mTOR) levels in RAAFCs were determined by Western blotting. Animals with the drug mesh had an intimal area index of 4.87% ± 0.98%, significantly lower than that of the blank group (14.21% ± 2.56%) or the PPC group (15.03% ± 2.35%, both P < .05). The sirolimus mesh markedly suppressed MMP-2 and MMP-9 expression, decreased PCNA-positive cell numbers, inhibited RAAFC migration, and reduced phospho-mTOR levels. Our data suggest that the sirolimus-eluting PPC mesh might be potentially applied for the management of grafting stenosis.
Collapse
Affiliation(s)
- Hourong Sun
- 1 Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chuan-Zhen Liu
- 1 Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chunxiao Liu
- 1 Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Mengmeng Tang
- 1 Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Guangqing Cao
- 1 Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qiuwang Zhang
- 2 Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Xinghua Gu
- 1 Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
307
|
Amphotericin B-loaded nanoparticles for local treatment of cutaneous leishmaniasis. Drug Deliv Transl Res 2018; 9:76-84. [DOI: 10.1007/s13346-018-00603-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
308
|
In-situ forming gels containing fluorometholone-loaded polymeric nanoparticles for ocular inflammatory conditions. Colloids Surf B Biointerfaces 2018; 175:365-374. [PMID: 30554015 DOI: 10.1016/j.colsurfb.2018.11.065] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 12/13/2022]
Abstract
Thermosensitive gels have been developed and optimized in such a way that they become gels at corneal temperature and with a viscosity that allows the adequate release of the Fluorometholone (FMT)-loaded PLGA nanoparticles (NPs) in order to improve ocular anti-inflammatory efficacy against a commercial formulation. It has been shown that gels avoid burst release of the drug in the first hours with a slow and increasing profile after administration. NPs have maintained their average size and spherical shape within the gels as confirmed by transmission electron microscopy (TEM). In turn, the in-situ gelling of the formulations allows the administration in eye drops dosage form due to its state of sol at temperatures below 25 °C. Ocular tolerance studies have shown that no formulation causes eye irritation. The administration of the developed formulations has improved the precorneal residence time reflected in the ocular bioavailability, where deep tissues as aqueous humour and crystalline were reached. In conclusion, the use of thermosensitive gels for the topical application of NPs has demonstrated their effectiveness in the acute and preventive treatment of ocular inflammatory conditions.
Collapse
|
309
|
Kumar P, Choonara YE, du Toit LC, Singh N, Pillay V. In Vitro and In Silico Analyses of Nicotine Release from a Gelisphere-Loaded Compressed Polymeric Matrix for Potential Parkinson's Disease Interventions. Pharmaceutics 2018; 10:pharmaceutics10040233. [PMID: 30445765 PMCID: PMC6320845 DOI: 10.3390/pharmaceutics10040233] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/07/2018] [Accepted: 11/12/2018] [Indexed: 01/21/2023] Open
Abstract
This study aimed to develop a prolonged-release device for the potential site-specific delivery of a neuroprotective agent (nicotine). The device was formulated as a novel reinforced crosslinked composite polymeric system with the potential for intrastriatal implantation in Parkinson's disease interventions. Polymers with biocompatible and bioerodible characteristics were selected to incorporate nicotine within electrolyte-crosslinked alginate-hydroxyethylcellulose gelispheres compressed within a release rate-modulating external polymeric matrix, comprising either hydroxypropylmethylcellulose (HPMC), polyethylene oxide (PEO), or poly(lactic-co-glycolic) acid (PLGA) to prolong nicotine release. The degradation and erosion studies showed that the produced device had desirable robustness with the essential attributes for entrapping drug molecules and retarding their release. Zero-order drug release was observed over 50 days from the device comprising PLGA as the external matrix. Furthermore, the alginate-nicotine interaction, the effects of crosslinking on the alginate-hydroxyethycellulose (HEC) blend, and the effects of blending PLGA, HPMC, and PEO on device performance were mechanistically elucidated using molecular modelling simulations of the 3D structure of the respective molecular complexes to predict the molecular interactions and possible geometrical orientation of the polymer morphologies affecting the geometrical preferences. The compressed polymeric matrices successfully retarded the release of nicotine over several days. PLGA matrices offered minimal rates of matrix degradation and successfully retarded nicotine release, leading to the achieved zero-order release for 50 days following exposure to simulated cerebrospinal fluid (CSF).
Collapse
Affiliation(s)
- Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| | - Lisa C du Toit
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| | - Neha Singh
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| | - Viness Pillay
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| |
Collapse
|
310
|
Wan S, Zhang L, Quan Y, Wei K. Resveratrol-loaded PLGA nanoparticles: enhanced stability, solubility and bioactivity of resveratrol for non-alcoholic fatty liver disease therapy. ROYAL SOCIETY OPEN SCIENCE 2018; 5:181457. [PMID: 30564426 PMCID: PMC6281916 DOI: 10.1098/rsos.181457] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/16/2018] [Indexed: 05/25/2023]
Abstract
Resveratrol (3, 4', 5-trihydroxy-trans-stilbene, RSV), a nutraceutical, has recently attracted lots of attention because of its outstanding pharmacological potential. The effects of RSV on non-alcoholic fatty liver disease (NAFLD) remain inconclusive, although a wealth of research has been done. The major obstacle presented was RSV's poor bioavailability due to its poor aqueous solubility, chemical instability and intestinal metabolism. In this study, nanotechnology was used to encapsulate RSV to enhance its stability, water solubility and bioactivity, which can be used to treat NAFLD by HepG2 hepatocytes-induced in vitro. RSV-loaded poly (d, l-lactide-co-glycolide acid) (PLGA) nanoparticles (RSV-PLGA-NPs) were prepared according to an oil/water (O/W) emulsion technique. The RSV-PLGA-NPs were of spherical morphology with an average size of 176.1 nm and a negative charge of -22.6 mV. These nanoparticles exhibited remarkable encapsulation efficiency (EE%) (97.25%) and drug loading (14.9%) for RSV. A sustained RSV release from RSV-PLGA-NPs could be achieved especially in acidic conditions when simulating transporting through the gastrointestinal tract. In addition, these nanoparticles were stable enough to store at 4°C for a least six months with unchanged EE%. Moreover, RSV-PLGA-NPs were more efficient in alleviating lipogenesis, promoting lipolysis and reducing hepatocellular proliferation than free RSV due to its improved stability, water solubility and bioactivity. These findings indicated that the RSV-PLGA-NPs provided superb and stable drug delivery with small particle size, high capsulation efficiency, well-controlled drug release, which greatly enhanced the stability, water solubility and bioactivity. Besides, the discovery that the inhibitory effect of RSV-PLGA-NPs on hepatocellular proliferation and lipid accumulation in steatotic HepG2 cells may provide a new way to study the mechanism of NAFLD. Therefore, RSV-PLGA-NPs have a promising potential for NAFLD therapy.
Collapse
Affiliation(s)
- Shuqian Wan
- School of Biological Science and Engineering, South China University of Technology, Guangzhou 510640, People's Republic of China
| | - Long Zhang
- Wenzhou Institute of Biomaterials and Engineering, CAS, Wenzhou, Zhejiang 325011, People's Republic of China
| | - Yunyun Quan
- Wenzhou Institute of Biomaterials and Engineering, CAS, Wenzhou, Zhejiang 325011, People's Republic of China
| | - Kun Wei
- School of Biological Science and Engineering, South China University of Technology, Guangzhou 510640, People's Republic of China
- Wenzhou Institute of Biomaterials and Engineering, CAS, Wenzhou, Zhejiang 325011, People's Republic of China
| |
Collapse
|
311
|
A molecular dynamics simulation study on the mechanism of loading of gemcitabine and camptothecin in poly lactic-co-glycolic acid as a nano drug delivery system. J Mol Liq 2018. [DOI: 10.1016/j.molliq.2018.08.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
312
|
Polyester based nanovehicles for siRNA delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:1006-1015. [DOI: 10.1016/j.msec.2018.05.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 02/12/2018] [Accepted: 05/07/2018] [Indexed: 12/18/2022]
|
313
|
Zhou Z, Yao Q, Li L, Zhang X, Wei B, Yuan L, Wang L. Antimicrobial Activity of 3D-Printed Poly(ε-Caprolactone) (PCL) Composite Scaffolds Presenting Vancomycin-Loaded Polylactic Acid-Glycolic Acid (PLGA) Microspheres. Med Sci Monit 2018; 24:6934-6945. [PMID: 30269152 PMCID: PMC6178870 DOI: 10.12659/msm.911770] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The aim of this study was to design and test a novel composite scaffold with antibacterial efficacy for treating bone infections using a three-dimensional (3D) printed poly(ε-caprolactone) (PCL) scaffold coated with polydopamine (PDA) for the adsorption of polylactic acid-glycolic acid (PLGA) microspheres loaded with vancomycin. MATERIAL AND METHODS Vancomycin-loaded PLGA microspheres were produced by the double-emulsion method, and microsphere morphology, drug-loading dosage, encapsulation efficiency, average diameter, and release characteristics were examined. Composite scaffolds were prepared by adsorption of the microspheres on PDA-coated, 3D-printed PCL scaffolds, and scaffold morphology, biocompatibility, vancomycin release, and antibacterial efficacy were evaluated. RESULTS The vancomycin-loaded microspheres were smooth, round, uniform in size, and had no adhesion phenomenon, and exhibited sustained release of vancomycin from the microspheres for more than 4 weeks. Upon modification with PDA, the PCL scaffold changed from white to black, and after microsphere adsorption, dot-like white particles were seen. On scanning electron microscopy, PDA particles were observed on the PCL/PDA composite scaffolds, and PLGA microspheres were evenly dispersed over the PDA coating on the PCL/PDA/PLGA composite scaffolds. Cell viability assays showed that the adhesion and proliferation of rabbit bone mesenchymal stem cells were greater on the PCL/PDA scaffolds than on unmodified PCL scaffolds. Microsphere adsorption had no significant effect on cell proliferation. In vitro release of vancomycin from the composite scaffolds was observed for more than 4 weeks, and observation of the inhibition zone on agar plates of Staphylococcus aureus showed that the scaffolds maintained their antibacterial effect for more than 4 weeks. CONCLUSIONS The 3D-printed, PDA-coated PCL scaffold carrying vancomycin-loaded PLGA microspheres exhibited good biocompatibility and a sustained antibacterial effect in vitro.
Collapse
Affiliation(s)
- Zhi Zhou
- Department of Orthopedics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (mainland)
| | - Qingqiang Yao
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China (mainland).,Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Lan Li
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, Jiangsu, China (mainland).,College of Biological and Pharmaceutical Engineering, Nanjing University of Technology, Nanjing, Jiangsu, China (mainland)
| | - Xin Zhang
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, Jiangsu, China (mainland).,College of Biological and Pharmaceutical Engineering, Nanjing University of Technology, Nanjing, Jiangsu, China (mainland)
| | - Bo Wei
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China (mainland).,Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Li Yuan
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Liming Wang
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China (mainland).,Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
314
|
Synergistic Effects of Controlled-Released BMP-2 and VEGF from nHAC/PLGAs Scaffold on Osteogenesis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3516463. [PMID: 30345299 PMCID: PMC6174819 DOI: 10.1155/2018/3516463] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 07/12/2018] [Accepted: 09/04/2018] [Indexed: 01/18/2023]
Abstract
Tissue engineering bones take great advantages in massive bone defect repairing; under the induction of growth factors, seed cells differentiate into osteoblasts, and the scaffold materials gradually degrade and are replaced with neogenetic bones, which simulates the actual pathophysiological process of bone regeneration. However, mechanism research is required and further developed to instruct elements selection and optimization. In the present study, we prepared vascular endothelial growth factor/bone morphogenetic protein-2- nanohydroxyapatite/collagen (VEGF/ BMP-2- nHAC/ PLGAs) scaffolds and inoculated mouse MC3T3-E1 preosteoblasts to detect osteogenic indexes and activation of related signaling pathways. The hypothesis is to create a three-dimensional environment that simulates bone defect repairing, and p38 mitogen-activated kinase (p38) inhibitor was applied and osterix shRNA was transferred into mouse MC3T3-E1 preosteoblasts to further investigate the molecular mechanism of crosstalk between BMP-2 and VEGF. Our results demonstrated the following: (1) BMP-2 and VEGF were sustainably released from PLGAs microspheres. (2) nHAC/PLGAs scaffold occupied a three-dimensional porous structure and has excellent physical properties. (3) MC3T3-E1 cells proliferated and differentiated well in the scaffold. (4) Osteogenic differentiation related factors expression of VEGF/BMP-2 loaded scaffold was obviously higher than that of other groups; p38 inhibitor SB203580 decreased the nucleus/cytoplasm ratio of osterix expression. To conclude, the active artificial bone we prepared could provide a favorable growth space for MC3T3-E1 cells, and osteogenesis and maturation reinforced by simultaneous VEGF and BMP-2 treatment may be mainly through the activation of the p38 MAPK pathway to promote nuclear translocation of osterix protein.
Collapse
|
315
|
PLGA: From a classic drug carrier to a novel therapeutic activity contributor. J Control Release 2018; 289:10-13. [PMID: 30244137 DOI: 10.1016/j.jconrel.2018.09.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 11/21/2022]
Abstract
Poly(lactic-co-glycolic acid) (PLGA) is well known for its biocompatibility and minimal toxicity. It is one of the most promising biodegradable polymeric drug delivery systems able to get endorsement from regulatory bodies to enter market. For many decades, PLGA has been functioning as an excipient, which by definition is pharmaceutically inert at a given dose of formulation. Lactate (one of the hydrolysis products of PLGA) has a key role in biochemical pathways and could improve physiological activities in certain illnesses by exerting therapeutic effects such as angiogenesis and promotion of healing. These activities, however, depend on the released amounts and metabolic clearance of lactate and route of formulation delivery. In the current commentary, along with several key notes on the lactate interactions, we would like to inform the PLGA research community that lactate (resulting from local delivery of physiologically significant amount of PLGA) may positively or negatively affect therapeutic efficacy of certain drugs. Hence, the excipient role of PLGA may be investigated for its potential pharmacological contributions in some biomedical applications.
Collapse
|
316
|
Zhang Y, Zhao J, Sun J, Huang L, Li Q. Targeting lung cancer initiating cells by all-trans retinoic acid-loaded lipid-PLGA nanoparticles with CD133 aptamers. Exp Ther Med 2018; 16:4639-4649. [PMID: 30542415 PMCID: PMC6257334 DOI: 10.3892/etm.2018.6762] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
Lung cancer initiating cells represent a specific subpopulation of lung cancer cells, which significantly contribute to the initiation, metastasis and recurrence of lung cancer. CD133, initially considered a marker of stem cells, is now considered as a marker for lung cancer initiating cells. All-trans retinoic acid (RA) has been demonstrated to cause the differentiation, inhibition of proliferation, and apoptosis of cancer cells and cancer initiating cells. However, there have been no reports on the activity of RA against lung cancer initiating cells. In the present study, the activity of RA against lung cancer initiating cells was investigated by determining the cytotoxicity, and performing a tumorsphere assay and flow cytometry-based analysis. In addition, to promote the therapeutic effect of RA in CD133+ lung cancer initiating cells, RA-loaded lipid poly(lactic-co-glycolic acid) (PLGA) nanoparticles with CD133 aptamers (RA-LPNPs-CD133) were developed. The activity of RA and RA-LPNPs-CD133 against lung cancer initiating cells was also investigated. RA-LPNPs-CD133 had a size of 129.9 nm, and exhibited sustained release of RA during the 144-h period. For the first time, to the best of our knowledge, the present study demonstrated that RA exerted potent activity towards CD133+ lung cancer initiating cells. The results also showed that RA-LPNPs-CD133 efficiently and specifically promoted the delivery of RA to CD133+ lung cancer initiating cells, exhibiting superior inhibitory effects against CD133+ lung cancer initiating cells compared with non-targeted nanoparticles and RA. To the best of our knowledge, the present study is the first to report the promotion of RA delivery via nanoparticles to lung cancer initiating cells and achievement of a superior inhibitory effect against lung cancer initiating cells by the utilization of CD133 aptamers. Therefore, RA-LPNPs-CD133 represents a promising tool for the elimination of lung cancer initiating cells.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| | - Juan Zhao
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| | - Jing Sun
- Department of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
| | - Lu Huang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| | - Qingfeng Li
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| |
Collapse
|
317
|
Peña Icart L, Fernandes dos Santos E, Agüero Luztonó L, Zaldívar Silva D, Andrade L, Lopes Dias M, Trambaioli da Rocha e Lima LM, Gomes de Souza F. Paclitaxel-Loaded PLA/PEG/Magnetite Anticancer and Hyperthermic Agent Prepared From Materials Obtained by the Ugi's Multicomponent Reaction. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/masy.201800094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Luis Peña Icart
- Centro de Ciências e Saúde, faculdade de farmácia; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
- Centro de biomateriais (BIOMAT); Universidade da Habana; Havana Cuba
- Instituto de Macromoléculas: Professora Eloisa Mano; Universidade Federal de Rio de Janeiro; Rio de Janeiro Brazil
| | | | | | | | - Leonardo Andrade
- Lab. de Biomineralização; Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - Marcos Lopes Dias
- Programa de Engenharia Civil, COPPE; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | | | - Fernando Gomes de Souza
- Instituto de Macromoléculas: Professora Eloisa Mano; Universidade Federal de Rio de Janeiro; Rio de Janeiro Brazil
- Programa de Engenharia Civil, COPPE; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| |
Collapse
|
318
|
Kharel S, Gautam A, Dickescheid A, Loo SCJ. Hollow Microparticles as a Superior Delivery System over Solid Microparticles for the Encapsulation of Peptides. Pharm Res 2018; 35:185. [PMID: 30073514 DOI: 10.1007/s11095-018-2461-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 07/15/2018] [Indexed: 12/27/2022]
|
319
|
Gonzalez-Pizarro R, Silva-Abreu M, Calpena AC, Egea MA, Espina M, García ML. Development of fluorometholone-loaded PLGA nanoparticles for treatment of inflammatory disorders of anterior and posterior segments of the eye. Int J Pharm 2018; 547:338-346. [DOI: 10.1016/j.ijpharm.2018.05.050] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/12/2018] [Accepted: 05/21/2018] [Indexed: 01/08/2023]
|
320
|
Kavanagh ON, Albadarin AB, Croker DM, Healy AM, Walker GM. Maximising success in multidrug formulation development: A review. J Control Release 2018; 283:1-19. [DOI: 10.1016/j.jconrel.2018.05.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/18/2018] [Accepted: 05/19/2018] [Indexed: 12/20/2022]
|
321
|
Segura-Ibarra V, Wu S, Hassan N, Moran-Guerrero JA, Ferrari M, Guha A, Karmouty-Quintana H, Blanco E. Nanotherapeutics for Treatment of Pulmonary Arterial Hypertension. Front Physiol 2018; 9:890. [PMID: 30061840 PMCID: PMC6055049 DOI: 10.3389/fphys.2018.00890] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/20/2018] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating and fatal chronic lung disease. While current pharmacotherapies have improved patient quality of life, PAH drugs suffer from limitations in the form of short-term pharmacokinetics, instability, and poor organ specificity. Traditionally, nanotechnology-based delivery strategies have proven advantageous at increasing both circulation lifetimes of chemotherapeutics and accumulation in tumors due to enhanced permeability through fenestrated vasculature. Importantly, increased nanoparticle (NP) accumulation in diseased tissues has been observed pre-clinically in pathologies characterized by endothelial dysfunction and remodeled vasculature, including myocardial infarction and heart failure. Recently, this phenomenon has also been observed in preclinical models of PAH, leading to the exploration of NP-based drug delivery as a therapeutic modality in PAH. Herein, we discussed the advantages of NPs for efficacious treatment of PAH, including heightened therapeutic delivery to diseased lungs for increased drug bioavailability, as well as highlighted innovative nanotherapeutic approaches for PAH.
Collapse
Affiliation(s)
- Victor Segura-Ibarra
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States.,Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey, Mexico
| | - Suhong Wu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Nida Hassan
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States.,McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jose A Moran-Guerrero
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States.,Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey, Mexico
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States.,Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Ashrith Guha
- Department of Cardiology, Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, Houston, TX, United States.,Houston Methodist J.C. Walter Jr. Transplant Center, Houston Methodist Hospital, Houston, TX, United States
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States.,Department of Cardiology, Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
322
|
Wan X, Pei W, Shahzad KA, Zhang L, Song S, Jin X, Wang L, Zhao C, Shen C. A Tolerogenic Artificial APC Durably Ameliorates Experimental Autoimmune Encephalomyelitis by Directly and Selectively Modulating Myelin Peptide–Autoreactive CD4+and CD8+T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:1194-1210. [DOI: 10.4049/jimmunol.1800108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 06/19/2018] [Indexed: 01/19/2023]
|
323
|
Zhou J, Sun J, Chen H, Peng Q. Promoted delivery of salinomycin sodium to lung cancer cells by dual targeting PLGA hybrid nanoparticles. Int J Oncol 2018; 53:1289-1300. [PMID: 30015824 DOI: 10.3892/ijo.2018.4474] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 06/05/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Jie Zhou
- Department of Pulmonary Medicine, Minhang Hospital, Zhongshan Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Jin Sun
- Department of Pharmacy, The Naval Medical University, Shanghai 200433, P.R. China
| | - Huaiwen Chen
- Center of Clinical and Translational Medicine, Shanghai Changhai Hospital, The Naval Medical University, Shanghai 200433, P.R. China
| | - Qing Peng
- Department of Pulmonary Medicine, Minhang Hospital, Zhongshan Hospital, Fudan University, Shanghai 201199, P.R. China
| |
Collapse
|
324
|
Huang X, Wang D, Yuan Z, Xie W, Wu Y, Li R, Zhao Y, Luo D, Cen L, Chen B, Wu H, Xu H, Sheng X, Zhang M, Zhao L, Yin L. A Fully Biodegradable Battery for Self-Powered Transient Implants. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1800994. [PMID: 29806124 DOI: 10.1002/smll.201800994] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/13/2018] [Indexed: 05/06/2023]
Abstract
Biodegradable transient devices represent an emerging type of electronics that could play an essential role in medical therapeutic/diagnostic processes, such as wound healing and tissue regeneration. The associated biodegradable power sources, however, remain as a major challenge toward future clinical applications, as the demonstrated electrical stimulation and sensing functions are limited by wired external power or wireless energy harvesters via near-field coupling. Here, materials' strategies and fabrication schemes that enable a high-performance fully biodegradable magnesium-molybdenum trioxide battery as an alternative approach for an in vivo on-board power supply are reported. The battery can deliver a stable high output voltage as well as prolonged lifetime that could satisfy requirements of representative implantable electronics. The battery is fully biodegradable and demonstrates desirable biocompatibility. The battery system provides a promising solution to advanced energy harvesters for self-powered transient bioresorbable implants as well as eco-friendly electronics.
Collapse
Affiliation(s)
- Xueying Huang
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Tsinghua University, Beijing, 100084, P. R. China
| | - Dan Wang
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Tsinghua University, Beijing, 100084, P. R. China
| | - Zhangyi Yuan
- Department of Electronic Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Wensheng Xie
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Tsinghua University, Beijing, 100084, P. R. China
| | - Yixin Wu
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Tsinghua University, Beijing, 100084, P. R. China
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Rongfeng Li
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Tsinghua University, Beijing, 100084, P. R. China
| | - Yu Zhao
- Department of Electronic Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Deng Luo
- Department of Electronic Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Liang Cen
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Tsinghua University, Beijing, 100084, P. R. China
| | - Binbin Chen
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Tsinghua University, Beijing, 100084, P. R. China
| | - Hui Wu
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Tsinghua University, Beijing, 100084, P. R. China
| | - Hangxun Xu
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Xing Sheng
- Department of Electronic Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Milin Zhang
- Department of Electronic Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Lingyun Zhao
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Tsinghua University, Beijing, 100084, P. R. China
| | - Lan Yin
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
325
|
Pei W, Wan X, Shahzad KA, Zhang L, Song S, Jin X, Wang L, Zhao C, Shen C. Direct modulation of myelin-autoreactive CD4 + and CD8 + T cells in EAE mice by a tolerogenic nanoparticle co-carrying myelin peptide-loaded major histocompatibility complexes, CD47 and multiple regulatory molecules. Int J Nanomedicine 2018; 13:3731-3750. [PMID: 29983566 PMCID: PMC6027825 DOI: 10.2147/ijn.s164500] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Purpose Numerous nanomaterials have been reported in the treatment of multiple sclerosis or experimental autoimmune encephalomyelitis (EAE). But most of these nanoscale therapeutics deliver myelin antigens together with toxins or cytokines and underlay the cellular uptake and induction of tolerogenic antigen-presenting cells by which they indirectly induce T cell tolerance. This study focuses on the on-target and direct modulation of myelin-autoreactive T cells and combined use of multiple regulatory molecules by generating a tolerogenic nanoparticle. Materials and methods Poly(lactic-co-glycolic acid) nanoparticles (PLGA-NPs) were fabricated by co-coupling MOG40–54/H-2Db-Ig dimer, MOG35–55/I-Ab multimer, anti-Fas, PD-L1-Fc and CD47-Fc and encapsulating transforming growth factor-β1. The resulting 217 nm tolerogenic nanoparticles (tNPs) were administered intravenously into MOG35–55 peptide-induced EAE mice, which was followed by the investigation of therapeutic outcomes and the in vivo mechanism. Results Four infusions of the tNPs durably ameliorated EAE with a marked reduction of clinical score, neuroinflammation and demyelination. They were distributed in secondary lymphoid tissues, various organs and brain after intravenous injection, with retention over 36 h, and made contacts with CD4+ and CD8+ T cells. Two injections of the tNPs markedly decreased the MOG35–55-reactive Th1 and Th17 cells and MOG40–55-reactive Tc1 and Tc17 cells, increased regulatory T cells, inhibited T cell proliferation and elevated T cell apoptosis in spleen. Transforming growth factor-β1 and interleukin-10 were upregulated in the homogenates of central nervous system and supernatant of spleen cells. Conclusion Our data suggest a novel therapeutic nanoparticle to directly modulate autoreactive T cells by surface presentation of multiple ligands and paracrine release of cytokine in the antigen-specific combination immunotherapy for T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Weiya Pei
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Xin Wan
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Khawar Ali Shahzad
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Lei Zhang
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Shilong Song
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Xiaoxiao Jin
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Limin Wang
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Chen Zhao
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Chuanlai Shen
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| |
Collapse
|
326
|
Askin S, Zhao M, Gonçalves AD, Gaisford S, Craig DQM. The Development of Quasi-isothermal Calorimetry for the Measurement of Drug–Polymer Miscibility and Crystallization Kinetics: Olanzapine-Loaded PLGA Microparticles. Mol Pharm 2018; 15:3332-3342. [DOI: 10.1021/acs.molpharmaceut.8b00364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Sean Askin
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Min Zhao
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Andrea D. Gonçalves
- DPDD Drug Delivery, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Simon Gaisford
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Duncan Q. M. Craig
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| |
Collapse
|
327
|
Abstract
Most diseases and disorders of the brain require long-term therapy and a constant supply of drugs. Implantable drug-delivery systems provide long-term, sustained drug delivery in the brain. The present review discusses different type of implantable systems such as solid implants, in situ forming implants, in situ forming microparticles, depot formulations, polymeric-lipid implants, sucrose acetate isobutyrate and N-stearoyl L-alanine methyl ester systems for continuous drug delivery into brain for various brain diseases including glioblastomas, medulloblastoma, epilepsy, stroke, schizophrenia and Alzheimer's diseases. Implantable neural probes and microelectrode array systems for brain are also discussed in brief.
Collapse
|
328
|
Su J, Liu G, Lian Y, Kamal Z, Que X, Qiu Y, Qiu M. Preparation and characterization of erythrocyte membrane cloaked PLGA/arsenic trioxide nanoparticles and evaluation of their in vitro anti-tumor effect. RSC Adv 2018; 8:20068-20076. [PMID: 35541656 PMCID: PMC9080777 DOI: 10.1039/c8ra01417e] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/01/2018] [Indexed: 12/30/2022] Open
Abstract
Arsenic trioxide (ATO) is used for acute promyelocytic leukemia (APL) that is resistant to all-trans-retinoic acid, but its direct intravenous injection sometimes induces severe toxic side effects. Here, we developed a delivery system of red blood cell membrane (RBCM) cloaked poly (lactic-co-glycolic) acid (PLGA)-ATO nanoparticles (RPANs) to reduce the toxicity. PLGA was used to entrap the ATO, and the PLGA-ATO nanoparticles (PANs) were prepared by the emulsification method. Then RBCMs were employed to cloak the PANs using ultrasonication, to develop the RPANs delivery system. The prepared RPANs had a uniform size of around 233.6 nm with an obvious core-shell structure, as observed by TEM. The completeness of the membrane proteins was confirmed by SDS-PAGE and an in vitro release time of 65 h was determined for the RPANs. The RPANs also exhibited low cytotoxicity against the 293k kidney cell line (84.6% cell viability rate), which suggested that the ATO toxicity was reduced by RBCM cloaking. Moreover, the anti-tumor effects of the RPANs against the HL60 cell line were comparable to those of ATO solution. Our study demonstrated that the RPANs system has anti-tumor potential and could be developed into a safe and sustained release delivery system for ATO.
Collapse
Affiliation(s)
- Jing Su
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
| | - Geyi Liu
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
| | - Yumei Lian
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
| | - Zul Kamal
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
- Department of Pharmacy, Shaheed Benazir Bhutto University Sheringal Dir (Upper) 18000 Khyber Pakhtunkhwa Pakistan
| | - Xiao Que
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
| | | | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
| |
Collapse
|
329
|
Zhang P, Xia J, Luo S. Generation of Well-Defined Micro/Nanoparticles via Advanced Manufacturing Techniques for Therapeutic Delivery. MATERIALS 2018; 11:ma11040623. [PMID: 29670013 PMCID: PMC5951507 DOI: 10.3390/ma11040623] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/08/2018] [Accepted: 04/11/2018] [Indexed: 12/15/2022]
Abstract
Micro/nanoparticles have great potentials in biomedical applications, especially for drug delivery. Existing studies identified that major micro/nanoparticle features including size, shape, surface property and component materials play vital roles in their in vitro and in vivo applications. However, a demanding challenge is that most conventional particle synthesis techniques such as emulsion can only generate micro/nanoparticles with a very limited number of shapes (i.e., spherical or rod shapes) and have very loose control in terms of particle sizes. We reviewed the advanced manufacturing techniques for producing micro/nanoparticles with precisely defined characteristics, emphasizing the use of these well-controlled micro/nanoparticles for drug delivery applications. Additionally, to illustrate the vital roles of particle features in therapeutic delivery, we also discussed how the above-mentioned micro/nanoparticle features impact in vitro and in vivo applications. Through this review, we highlighted the unique opportunities in generating controllable particles via advanced manufacturing techniques and the great potential of using these micro/nanoparticles for therapeutic delivery.
Collapse
Affiliation(s)
- Peipei Zhang
- Department of Material Processing and Controlling, School of Mechanical Engineering & Automation, Beihang University, Beijing 100191, China.
| | - Junfei Xia
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA.
| | - Sida Luo
- Department of Material Processing and Controlling, School of Mechanical Engineering & Automation, Beihang University, Beijing 100191, China.
| |
Collapse
|
330
|
Belletti D, Grabrucker AM, Pederzoli F, Menrath I, Vandelli MA, Tosi G, Duskey TJ, Forni F, Ruozi B. Hybrid nanoparticles as a new technological approach to enhance the delivery of cholesterol into the brain. Int J Pharm 2018; 543:300-310. [PMID: 29608954 DOI: 10.1016/j.ijpharm.2018.03.061] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 12/25/2022]
Abstract
Restoration of the Chol homeostasis in the Central Nervous System (CNS) could be beneficial for the treatment of Huntington's Disease (HD), a progressive, fatal, adult-onset, neurodegenerative disorder. Unfortunately, Chol is unable to cross the blood-brain barrier (BBB), thus a novel strategy for a targeted delivery of Chol into the brain is highly desired. This article aims to investigate the production of hybrid nanoparticles composed by Chol and PLGA (MIX-NPs) modified with g7 ligand for BBB crossing. We described the impact of ratio between components (Chol and PLGA) and formulation process (nanoprecipitation or single emulsion process) on physico-chemical and structural characteristics, we tested MIX-NPs in vitro using primary hippocampal cell cultures evaluating possible toxicity, uptake, and the ability to influence excitatory synaptic receptors. Our results elucidated that both formulation processes produce MIX-NPs with a Chol content higher that 40%, meaning that Chol is a structural particle component and active compound at the same time. The formulation strategy impacted the architecture and reorganization of components leading to some differences in Chol availability between the two types of g7 MIX-NPs. Our results identified that both kinds of MIX-NPs are efficiently taken up by neurons, able to escape lysosomes and release Chol into the cells resulting in an efficient modification in expression of synaptic receptors that could be beneficial in HD.
Collapse
Affiliation(s)
- Daniela Belletti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Andreas Martin Grabrucker
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Bernal Institute, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| | - Francesca Pederzoli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Isabel Menrath
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Giovanni Tosi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Thomas Jason Duskey
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Flavio Forni
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Barbara Ruozi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
331
|
Priming the body to receive the therapeutic agent to redefine treatment benefit/risk profile. Sci Rep 2018; 8:4797. [PMID: 29556068 PMCID: PMC5859131 DOI: 10.1038/s41598-018-23140-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/06/2018] [Indexed: 12/16/2022] Open
Abstract
Many therapeutic agents offer a low useful dose (dose responsible for efficacy)/useless dose (dose eliminated or responsible for toxicity) ratio, mainly due to the fact that therapeutic agents must ensure in one single object all the functions required to deliver the treatment, which leads to compromises in their physico-chemical design. Here we introduce the concept of priming the body to receive the treatment by uncorrelating these functions into two distinct objects sequentially administered: a nanoprimer occupying transiently the main pathway responsible for therapeutic agent limited benefit/risk ratio followed by the therapeutic agent. The concept was evaluated for different nature of therapeutic agents: For nanomedicines we designed a liposomal nanoprimer presenting preferential hepatic accumulation without sign of acute toxicity. This nanoprimer was able to increase the blood bioavailability of nanomedicine correlated with a lower hepatic accumulation. Finally this nanoprimer markedly enhanced anti-tumor efficacy of irinotecan loaded liposomes in the HT-29 tumor model when compared to the nanomedicine alone. Then, for small molecules we demonstrated the ability of a cytochrome inhibitor loaded nanoprimer to increase efficacy of docetaxel treatment. These results shown that specific nanoprimers could be designed for each family of therapeutic agents to answer to their specific needs.
Collapse
|
332
|
Nakhlband A, Eskandani M, Omidi Y, Saeedi N, Ghaffari S, Barar J, Garjani A. Combating atherosclerosis with targeted nanomedicines: recent advances and future prospective. ACTA ACUST UNITED AC 2018; 8:59-75. [PMID: 29713603 PMCID: PMC5915710 DOI: 10.15171/bi.2018.08] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/21/2018] [Indexed: 12/19/2022]
Abstract
![]()
Introduction:
Cardiovascular diseases (CVDs) is recognized as the leading cause of mortality worldwide. The increasing prevalence of such disease demands novel therapeutic and diagnostic approaches to overcome associated clinical/social issues. Recent advances in nanotechnology and biological sciences have provided intriguing insights to employ targeted Nanomachines to the desired location as imaging, diagnosis, and therapeutic modalities. Nanomedicines as novel tools for enhanced drug delivery, imaging, and diagnosis strategies have shown great promise to combat cardiovascular diseases.
Methods:
In the current study, we intend to review the most recent studies on the nano-based strategies for improved management of CVDs.
Results:
A cascade of events results in the formation of atheromatous plaque and arterial stenosis. Furthermore, recent studies have shown that nanomedicines have displayed unique functionalities and provided de novo applications in the diagnosis and treatment of atherosclerosis.
Conclusion:
Despite some limitations, nanomedicines hold considerable potential in the prevention, diagnosis, and treatment of various ailments including atherosclerosis. Fewer side effects, amenable physicochemical properties and multi-potential application of such nano-systems are recognized through various investigations. Therefore, it is strongly believed that with targeted drug delivery to atherosclerotic lesions and plaque, management of onset and progression of disease would be more efficient than classical treatment modalities.
Collapse
Affiliation(s)
- Ailar Nakhlband
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazli Saeedi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Garjani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of pharmacology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
333
|
Hibbitts A, O'Leary C. Emerging Nanomedicine Therapies to Counter the Rise of Methicillin-Resistant Staphylococcus aureus. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E321. [PMID: 29473883 PMCID: PMC5849018 DOI: 10.3390/ma11020321] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/14/2018] [Accepted: 02/19/2018] [Indexed: 12/25/2022]
Abstract
In a recent report, the World Health Organisation (WHO) classified antibiotic resistance as one of the greatest threats to global health, food security, and development. Methicillin-resistant Staphylococcus aureus (MRSA) remains at the core of this threat, with persistent and resilient strains detectable in up to 90% of S. aureus infections. Unfortunately, there is a lack of novel antibiotics reaching the clinic to address the significant morbidity and mortality that MRSA is responsible for. Recently, nanomedicine strategies have emerged as a promising therapy to combat the rise of MRSA. However, these approaches have been wide-ranging in design, with few attempts to compare studies across scientific and clinical disciplines. This review seeks to reconcile this discrepancy in the literature, with specific focus on the mechanisms of MRSA infection and how they can be exploited by bioactive molecules that are delivered by nanomedicines, in addition to utilisation of the nanomaterials themselves as antibacterial agents. Finally, we discuss targeting MRSA biofilms using nano-patterning technologies and comment on future opportunities and challenges for MRSA treatment using nanomedicine.
Collapse
Affiliation(s)
- Alan Hibbitts
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
- Trinity Centre of Bioengineering, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.
- Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland.
| | - Cian O'Leary
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
- Trinity Centre of Bioengineering, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.
- Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland.
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
334
|
Yi Y, Lin G, Chen S, Liu J, Zhang H, Mi P. Polyester micelles for drug delivery and cancer theranostics: Current achievements, progresses and future perspectives. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 83:218-232. [DOI: 10.1016/j.msec.2017.10.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 12/14/2022]
|
335
|
Abu Ammar A, Gruber M, Martin P, Stern O, Jahshan F, Ertracht O, Sela E, Srouji S, Zussman E. Local delivery of mometasone furoate from an eluting endotracheal tube. J Control Release 2018; 272:54-61. [PMID: 29331580 DOI: 10.1016/j.jconrel.2018.01.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/06/2018] [Accepted: 01/08/2018] [Indexed: 12/15/2022]
Abstract
Laryngeal and tracheal morbidity is a common complication of endotracheal tube (ETT)-based airway management, and manifests as local irritation, inflammation, and edema. Systemic corticosteroids are commonly administered to manage these conditions; however, their efficacy is inadequate and limited by potential severe side effects. In the present study, a steroid delivery system for local therapy was developed to generate relatively high local drug concentrations and to improve drug efficacy. ETTs were coated with electrospun poly (lactic-co-glycolic acid) (PLGA) nanofibers loaded with mometasone furoate (MF), creating a microscale thick layer. MF exhibited sustained release from coated ETTs over 14days in vitro. An in vivo efficacy study in rats demonstrated the therapeutic benefit of MF-coated ETTs over bare ETTs, as measured by reduced laryngeal mucosal thickness and submucosal laryngeal edema. The fiber coating remained intact during tube intubation and extubation, demonstrating good adhesion to the tubes even after 24h in aqueous solution at 37°C. These findings demonstrate the potential of drug-loaded ETTs to revolutionize the standard of care for endotracheal intubation.
Collapse
Affiliation(s)
- Aiman Abu Ammar
- NanoEngineering Group, Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Maayan Gruber
- Department of Otolaryngology-Head and Neck Surgery, Galilee Medical Center, Nahariya 2210001, Israel; Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311502, Israel
| | - Patrick Martin
- NanoEngineering Group, Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Ohad Stern
- NanoEngineering Group, Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Forsan Jahshan
- Department of Otolaryngology-Head and Neck Surgery, Galilee Medical Center, Nahariya 2210001, Israel
| | - Offir Ertracht
- Eliachar Research Laboratory, Galilee Medical Center, Nahariya 2210001, Israel
| | - Eyal Sela
- Department of Otolaryngology-Head and Neck Surgery, Galilee Medical Center, Nahariya 2210001, Israel; Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311502, Israel
| | - Samer Srouji
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311502, Israel; Institute of Oral and Maxillofacial Surgery and Oral Medicine, Galilee Medical Center, Nahariya 2210001, Israel
| | - Eyal Zussman
- NanoEngineering Group, Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
336
|
Martins C, Sousa F, Araújo F, Sarmento B. Functionalizing PLGA and PLGA Derivatives for Drug Delivery and Tissue Regeneration Applications. Adv Healthc Mater 2018; 7. [PMID: 29171928 DOI: 10.1002/adhm.201701035] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/27/2017] [Indexed: 12/16/2022]
Abstract
Poly(lactic-co-glycolic) acid (PLGA) is one of the most versatile biomedical polymers, already approved by regulatory authorities to be used in human research and clinics. Due to its valuable characteristics, PLGA can be tailored to acquire desirable features for control bioactive payload or scaffold matrix. Moreover, its chemical modification with other polymers or bioconjugation with molecules may render PLGA with functional properties that make it the Holy Grail among the synthetic polymers to be applied in the biomedical field. In this review, the physical-chemical properties of PLGA, its synthesis, degradation, and conjugation with other polymers or molecules are revised in detail, as well as its applications in drug delivery and regeneration fields. A particular focus is given to successful examples of products already on the market or at the late stages of trials, reinforcing the potential of this polymer in the biomedical field.
Collapse
Affiliation(s)
- Cláudia Martins
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
| | - Flávia Sousa
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- ICBAS - Instituto Ciências Biomédicas Abel Salazar; Universidade do Porto; Rua de Jorge Viterbo Ferreira 228 4050-313 Porto Portugal
| | - Francisca Araújo
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde; Rua Central de Gandra 1317 4585-116 Gandra Portugal
| |
Collapse
|
337
|
Wang W, Fang K, Li MC, Chang D, Shahzad KA, Xu T, Zhang L, Gu N, Shen CL. A biodegradable killer microparticle to selectively deplete antigen-specific T cells in vitro and in vivo. Oncotarget 2017; 7:12176-90. [PMID: 26910923 PMCID: PMC4914277 DOI: 10.18632/oncotarget.7519] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/05/2016] [Indexed: 12/23/2022] Open
Abstract
The specific eradication of pathogenic T cells for the treatment of allograft rejections and autoimmune disorders without impairment of overall immune function is a fundamental goal. Here, cell-sized poly(lactic-co-glycolic acid) microparticles (PLGA MPs) were prepared as a scaffold to co-display the peptide/major histocompatibility complex (pMHC, target antigen) and anti-Fas monoclonal antibody (apoptosis-inducing molecule) for the generation of biodegradable killer MPs. Ovalbumin (OVA) antigen-targeted killer MPs significantly depleted OVA-specific CD8+ T cells in an antigen-specific manner, both in vitro and in OT-1 mice. After intravenous administration, the killer MPs predominantly accumulated in the liver, lungs, and gut of OT-1 mice with a retention time of up to 48 hours. The killing effects exerted by killer MPs persisted for 4 days after two injections. Moreover, the H-2Kb alloantigen-targeted killer MPs were able to eliminate low-frequency alloreactive T cells and prolong alloskin graft survival for 41.5 days in bm1 mice. Our data indicate that PLGA-based killer MPs are capable of specifically depleting pathogenic T cells, which highlights their therapeutic potential for treating allograft rejection and autoimmune disorders.
Collapse
Affiliation(s)
- Wei Wang
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, PR China
| | - Kun Fang
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, PR China
| | - Miao-Chen Li
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, PR China
| | - Di Chang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, PR China
| | - Khawar Ali Shahzad
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, PR China
| | - Tao Xu
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, PR China
| | - Lei Zhang
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, PR China
| | - Ning Gu
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, PR China
| | - Chuan-Lai Shen
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, PR China
| |
Collapse
|
338
|
Mangrio FA, Dwivedi P, Han S, Zhao G, Gao D, Si T, Xu RX. Characteristics of Artemether-Loaded Poly(lactic-co-glycolic) Acid Microparticles Fabricated by Coaxial Electrospray: Validation of Enhanced Encapsulation Efficiency and Bioavailability. Mol Pharm 2017; 14:4725-4733. [PMID: 29096443 DOI: 10.1021/acs.molpharmaceut.7b00862] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Artemether is one of the most effective drugs for the treatment of chloroquine-resistant and Plasmodium falciparum strains of malaria. However, its therapeutic potency is hindered by its poor bioavailability. To overcome this limitation, we have encapsulated artemether in poly(lactic-co-glycolic) acid (PLGA) core-shell microparticles (MPs) using the coaxial electrospray method. With optimized process parameters including liquid flow rates and applied electric voltages, experiments are systematically carried out to generate a stable cone-jet mode to produce artemether-loaded PLGA-MPs with an average size of 2 μm, an encapsulation efficiency of 78 ± 5.6%, and a loading efficiency of 11.7%. The in vitro release study demonstrates the sustained release of artemether from the core-shell structure in comparison with that of plain artemether and that of MPs produced by single-axial electrospray without any relevant cytotoxicity. The in vivo studies are performed to evaluate the pharmacokinetic characteristics of the artemether-loaded PLGA-MPs. Our study implies that artemether can be effectively encapsulated in a protective shell of PLGA for controlled release kinetics and enhanced oral bioavailability.
Collapse
Affiliation(s)
- Farhana Akbar Mangrio
- Department of Electronic Science and Technology, University of Science and Technology of China , Hefei 230027, China.,School of Engineering Science, University of Science and Technology of China , Hefei 230027, China
| | - Pankaj Dwivedi
- School of Engineering Science, University of Science and Technology of China , Hefei 230027, China
| | - Shuya Han
- School of Engineering Science, University of Science and Technology of China , Hefei 230027, China
| | - Gang Zhao
- Department of Electronic Science and Technology, University of Science and Technology of China , Hefei 230027, China
| | - Dayong Gao
- Department of Electronic Science and Technology, University of Science and Technology of China , Hefei 230027, China.,Department of Mechanical Engineering, University of Washington , Seattle, Washington 98195, United States
| | - Ting Si
- School of Engineering Science, University of Science and Technology of China , Hefei 230027, China
| | - Ronald X Xu
- School of Engineering Science, University of Science and Technology of China , Hefei 230027, China.,Department of Biomedical Engineering, The Ohio State University , Columbus, Ohio 43210, United States
| |
Collapse
|
339
|
Thulasidasan AKT, Retnakumari AP, Shankar M, Vijayakurup V, Anwar S, Thankachan S, Pillai KS, Pillai JJ, Nandan CD, Alex VV, Chirayil TJ, Sundaram S, Kumar GSV, Anto RJ. Folic acid conjugation improves the bioavailability and chemosensitizing efficacy of curcumin-encapsulated PLGA-PEG nanoparticles towards paclitaxel chemotherapy. Oncotarget 2017; 8:107374-107389. [PMID: 29296172 PMCID: PMC5746074 DOI: 10.18632/oncotarget.22376] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/25/2017] [Indexed: 12/18/2022] Open
Abstract
Nanoencapsulation has emerged as a novel strategy to enhance the pharmacokinetic and therapeutic potential of conventional drugs. Recent studies from our lab have established the efficacy of curcumin in sensitizing cervical cancer cells and breast cancer cells towards paclitaxel and 5-FU chemotherapy respectively. Factors that hinder the clinical use of curcumin as a sensitizer or therapeutic agent include its poor bioavailability and retention time. Earlier reports of improvement in bioavailability and retention of drugs upon nanoencapsulation have motivated us in developing various nanoformulations of curcumin, which were found to exhibit significant enhancement in bioavailability and retention time as assessed by our previous in vitro studies. Among the various formulations tested, curcumin-entrapped in PLGA-PEG nanoparticles conjugated to folic acid (PPF-curcumin) displayed maximum cell death. In the present study, we have demonstrated the efficacy of this formulation in augmenting the bioavailability and retention time of curcumin, in vivo, in Swiss albino mice. Further, the acute and chronic toxicity studies proved that the formulation is pharmacologically safe. We have also evaluated its potential in chemosensitizing cervical cancer cells to paclitaxel and have verified the results using cervical cancer xenograft model in NOD-SCID mice. Folic acid conjugation significantly enhanced the efficacy of curcumin in down-regulating various survival signals induced by paclitaxel in cervical cancer cells and have considerably improved its potential in inhibiting the tumor growth of cervical cancer xenografts. The non-toxic nature coupled with improved chemosensitization potential makes PPF-curcumin a promising candidate formulation for clinical trials.
Collapse
Affiliation(s)
- Arun Kumar T Thulasidasan
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Research Scholar, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Archana P Retnakumari
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Mohan Shankar
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Research Scholar, Manipal University, Manipal, Karnataka, India
| | - Vinod Vijayakurup
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Shabna Anwar
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Research Scholar, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Sanu Thankachan
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Kavya S Pillai
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Jisha J Pillai
- Division of Chemical Biology-Nano Drug Delivery Systems, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - C Devika Nandan
- Division of Chemical Biology-Nano Drug Delivery Systems, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Vijai V Alex
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Teena Jacob Chirayil
- Division of Chemical Biology-Nano Drug Delivery Systems, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Research Scholar, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Sankar Sundaram
- Department of Pathology, Government Medical College, Kottayam, Kerala, India
| | | | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
340
|
Mir M, Ahmed N, Rehman AU. Recent applications of PLGA based nanostructures in drug delivery. Colloids Surf B Biointerfaces 2017; 159:217-231. [DOI: 10.1016/j.colsurfb.2017.07.038] [Citation(s) in RCA: 325] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/06/2017] [Accepted: 07/16/2017] [Indexed: 12/12/2022]
|
341
|
Gao L, Xia L, Zhang R, Duan D, Liu X, Xu J, Luo L. Enhanced antitumor efficacy of poly(D,L-lactide-co-glycolide)-based methotrexate-loaded implants on sarcoma 180 tumor-bearing mice. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:3065-3075. [PMID: 29118572 PMCID: PMC5659257 DOI: 10.2147/dddt.s143942] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Purpose Methotrexate is widely used in chemotherapy for a variety of malignancies. However, severe toxicity, poor pharmacokinetics, and narrow safety margin of methotrexate limit its clinical application. The aim of this study was to develop sustained-release methotrexate-loaded implants and evaluate antitumor activity of the implants after intratumoral implantation. Materials and methods We prepared the implants containing methotrexate, poly(D,L-lactide-co-glycolide), and polyethylene glycol 4000 with the melt-molding technique. The implants were characterized with regards to drug content, morphology, in vitro, and in vivo release profiles. Differential scanning calorimetry (DSC) and Fourier transform infrared spectroscopy (FTIR) were carried out to investigate the physicochemical properties of the implants. Furthermore, the antitumor activity of the implants was tested in a sarcoma 180 mouse model. Results The implants were prepared as solid rods. Scanning electron microscopy images showed a smooth surface of the implant, suggesting that methotrexate was homogeneously dispersed in the polymeric matrix. The results of DSC and FTIR indicated that no significant interaction between methotrexate and the polymer was observed in the implants. Both in vitro and in vivo release profiles of the implants were characterized by burst release followed by sustained release of methotrexate. Intratumoral implantation of methotrexate-loaded implants could efficiently delay tumor growth. Moreover, an increase in the dose of implants led to a higher tumor suppression rate without additional systemic toxicity. Conclusion These results demonstrate that methotrexate-loaded implants had significant antitumor efficacy in a sarcoma 180 mouse model without dose-limiting side effects, and suggest that the implants could be potentially applied as an intratumoral delivery system to treat cancer.
Collapse
Affiliation(s)
- Li Gao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing.,School of Biological and Medical Engineering, Hefei University of Technology, Hefei
| | - Lunyang Xia
- Laboratory of Pharmaceutical Research, Anhui Zhongren Science and Technology Co., Ltd., Hefei, People's Republic of China
| | - Ruhui Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing
| | - Dandan Duan
- Laboratory of Pharmaceutical Research, Anhui Zhongren Science and Technology Co., Ltd., Hefei, People's Republic of China
| | - Xiuxiu Liu
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei
| | - Jianjian Xu
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing
| |
Collapse
|
342
|
Icart LP, Santos ERF, Agüero L, Andrade LR, de Souza CG, d´Avila LA, Zaldivar D, Dias ML. Paclitaxel-loaded PLA/PEG/fluorescein anticancer agent prepared by Ugi reaction. INT J POLYM MATER PO 2017. [DOI: 10.1080/00914037.2017.1378884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- L. P. Icart
- Laboratório de Biotecnologia Farmacêutica (pbiotech), Faculdade de farmácia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Edson R. F. Santos
- Centro de Tecnologia, COPPE, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - L. Agüero
- Instituto de Biomateriales (BIOMAT), Universidad de la Habana, Havana, Cuba
| | - Leonardo R. Andrade
- Laboratório de Biomineralização, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - C. G. de Souza
- Laboratório de Biocombustíveis, Escola de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - L. A. d´Avila
- Laboratório de Biocombustíveis, Escola de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - D. Zaldivar
- Instituto de Biomateriales (BIOMAT), Universidad de la Habana, Havana, Cuba
| | - M. L. Dias
- Instituto de Macromoléculas Professora Eloisa Mano (IMA), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
343
|
Reber LL, Starkl P, Balbino B, Sibilano R, Gaudenzio N, Rogalla S, Sensarn S, Kang D, Raghu H, Sokolove J, Robinson WH, Contag CH, Tsai M, Galli SJ. The tyrosine kinase inhibitor imatinib mesylate suppresses uric acid crystal-induced acute gouty arthritis in mice. PLoS One 2017; 12:e0185704. [PMID: 28982129 PMCID: PMC5628843 DOI: 10.1371/journal.pone.0185704] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 09/18/2017] [Indexed: 01/01/2023] Open
Abstract
Gouty arthritis is caused by the deposition of monosodium urate (MSU) crystals in joints. Despite many treatment options for gout, there is a substantial need for alternative treatments for patients unresponsive to current therapies. Tyrosine kinase inhibitors have demonstrated therapeutic benefit in experimental models of antibody-dependent arthritis and in rheumatoid arthritis in humans, but to date, the potential effects of such inhibitors on gouty arthritis has not been evaluated. Here we demonstrate that treatment with the tyrosine kinase inhibitor imatinib mesylate (imatinib) can suppress inflammation induced by injection of MSU crystals into subcutaneous air pouches or into the ankle joint of wild type mice. Moreover, imatinib treatment also largely abolished the lower levels of inflammation which developed in IL-1R1-/- or KitW-sh/W-sh mice, indicating that this drug can inhibit IL-1-independent pathways, as well as mast cell-independent pathways, contributing to pathology in this model. Imatinib treatment not only prevented ankle swelling and synovial inflammation when administered before MSU crystals but also diminished these features when administrated after the injection of MSU crystals, a therapeutic protocol more closely mimicking the clinical situation in which treatment occurs after the development of an acute gout flare. Finally, we also assessed the efficiency of local intra-articular injections of imatinib-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles in this model of acute gout. Treatment with low doses of this long-acting imatinib:PLGA formulation was able to reduce ankle swelling in a therapeutic protocol. Altogether, these results raise the possibility that tyrosine kinase inhibitors might have utility in the treatment of acute gout in humans.
Collapse
Affiliation(s)
- Laurent L. Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
- Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Immunology, Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Philipp Starkl
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
- Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, California, United States of America
| | - Bianca Balbino
- Department of Immunology, Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Paris, France
- Université Pierre et Marie Curie, Paris, France
| | - Riccardo Sibilano
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
- Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, California, United States of America
| | - Nicolas Gaudenzio
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
- Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, California, United States of America
| | - Stephan Rogalla
- Departments of Bioengineering, Radiology, and Pediatrics Division of Neonatology, Stanford University School of Medicine, Stanford, California, United States of America
- Molecular Imaging Program at Stanford, Stanford, California, United States of America
| | - Steven Sensarn
- Departments of Bioengineering, Radiology, and Pediatrics Division of Neonatology, Stanford University School of Medicine, Stanford, California, United States of America
- Molecular Imaging Program at Stanford, Stanford, California, United States of America
| | - Dongmin Kang
- Departments of Bioengineering, Radiology, and Pediatrics Division of Neonatology, Stanford University School of Medicine, Stanford, California, United States of America
- Molecular Imaging Program at Stanford, Stanford, California, United States of America
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Harini Raghu
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Geriatric Research Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Jeremy Sokolove
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Geriatric Research Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - William H. Robinson
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Geriatric Research Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Christopher H. Contag
- Departments of Bioengineering, Radiology, and Pediatrics Division of Neonatology, Stanford University School of Medicine, Stanford, California, United States of America
- Molecular Imaging Program at Stanford, Stanford, California, United States of America
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
- Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, California, United States of America
| | - Stephen J. Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
- Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
344
|
Huang N, Lu S, Liu XG, Zhu J, Wang YJ, Liu RT. PLGA nanoparticles modified with a BBB-penetrating peptide co-delivering Aβ generation inhibitor and curcumin attenuate memory deficits and neuropathology in Alzheimer's disease mice. Oncotarget 2017; 8:81001-81013. [PMID: 29113362 PMCID: PMC5655257 DOI: 10.18632/oncotarget.20944] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/26/2017] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by the formation of extracellular senile plaques and neuronal loss caused by amyloid β (Aβ) aggregates in the brains of AD patients. Conventional strategies failed to treat AD in clinical trials, partly due to the poor solubility, low bioavailability and ineffectiveness of the tested drugs to cross the blood-brain barrier (BBB). Moreover, AD is a complex, multifactorial neurodegenerative disease; one-target strategies may be insufficient to prevent the processes of AD. Here, we designed novel kind of poly(lactide-co-glycolic acid) (PLGA) nanoparticles by loading with Aβ generation inhibitor S1 (PQVGHL peptide) and curcumin to target the detrimental factors in AD development and by conjugating with brain targeting peptide CRT (cyclic CRTIGPSVC peptide), an iron-mimic peptide that targets transferrin receptor (TfR), to improve BBB penetration. The average particle size of drug-loaded PLGA nanoparticles and CRT-conjugated PLGA nanoparticles were 128.6 nm and 139.8 nm, respectively. The results of Y-maze and new object recognition test demonstrated that our PLGA nanoparticles significantly improved the spatial memory and recognition in transgenic AD mice. Moreover, PLGA nanoparticles remarkably decreased the level of Aβ, reactive oxygen species (ROS), TNF-α and IL-6, and enhanced the activities of super oxide dismutase (SOD) and synapse numbers in the AD mouse brains. Compared with other PLGA nanoparticles, CRT peptide modified-PLGA nanoparticles co-delivering S1 and curcumin exhibited most beneficial effect on the treatment of AD mice, suggesting that conjugated CRT peptide, and encapsulated S1 and curcumin exerted their corresponding functions for the treatment.
Collapse
Affiliation(s)
- Na Huang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.,Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan 750021, China
| | - Shuai Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiao-Ge Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Zhu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Yu-Jiong Wang
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan 750021, China
| | - Rui-Tian Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
345
|
Zhang L, Yao M, Yan W, Liu X, Jiang B, Qian Z, Gao Y, Lu XJ, Chen X, Wang QL. Delivery of a chemotherapeutic drug using novel hollow carbon spheres for esophageal cancer treatment. Int J Nanomedicine 2017; 12:6759-6769. [PMID: 28932119 PMCID: PMC5600264 DOI: 10.2147/ijn.s142916] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Low toxicity and high efficacy are the key factors influencing the real-world clinical applications of nanomaterial-assisted drug delivery. In this study, novel hollow carbon spheres (HCSs) with narrow size distribution were developed. In addition to demonstrating their ease of synthesis for large-scale production, we also demonstrated in vitro that the HCSs possessed high drug-loading capacity, lower cell toxicity, and optimal drug release profile at low pH, similar to the pH in the tumor microenvironment. The HCSs also displayed excellent immunocompatibility and could rapidly distribute themselves in the cytoplasm to escape lysosomal clearance. More importantly, the HCSs could efficiently deliver doxorubicin (a representative chemotherapeutic drug) to tumor sites, which resulted in significant inhibition of tumor growth in an esophageal xenograft cancer model. This also prolonged the circulation time and altered the biodistribution of the drug. In conclusion, this study revealed a novel drug delivery system for targeted tumor therapy.
Collapse
Affiliation(s)
| | - Mengchu Yao
- Department of Clinical Oncology.,Huai'an Key Laboratory of Esophageal Cancer Biobank
| | - Wei Yan
- Department of Gastroenterology
| | | | - Baofei Jiang
- Department of Gastrointestinal Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an
| | - Zhaoye Qian
- Department of Clinical Oncology.,Huai'an Key Laboratory of Esophageal Cancer Biobank
| | - Yong Gao
- Department of Clinical Oncology.,Huai'an Key Laboratory of Esophageal Cancer Biobank
| | - Xiao-Jie Lu
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | - Qi-Long Wang
- Department of Clinical Oncology.,Huai'an Key Laboratory of Esophageal Cancer Biobank.,Department of Central Laboratory
| |
Collapse
|
346
|
Bernocchi B, Carpentier R, Betbeder D. Nasal nanovaccines. Int J Pharm 2017; 530:128-138. [PMID: 28698066 DOI: 10.1016/j.ijpharm.2017.07.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 01/08/2023]
Abstract
Nasal administration of vaccines is convenient for the potential stimulation of mucosal and systemic immune protection. Moreover the easy accessibility of the intranasal route renders it optimal for pandemic vaccination. Nanoparticles have been identified as ideal delivery systems and adjuvants for vaccine application. Heterogeneous protocols have been used for animal studies. This complicates the understanding of the formulation influence on the immune response and the comparison of the different nanoparticles approaches developed. Moreover anatomical and immunological differences between rodents and humans provide an additional hurdle in the rational development of nasal nanovaccines. This review will give a comprehensive expertise of the state of the art in nasal nanovaccines in animals and humans focusing on the nanomaterial used.
Collapse
Affiliation(s)
- B Bernocchi
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France
| | - R Carpentier
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France.
| | - D Betbeder
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France; University of Artois, 62000 Arras, France
| |
Collapse
|
347
|
Localization of dexamethasone within dendritic core-multishell (CMS) nanoparticles and skin penetration properties studied by multi-frequency electron paramagnetic resonance (EPR) spectroscopy. Eur J Pharm Biopharm 2017; 116:94-101. [DOI: 10.1016/j.ejpb.2016.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 09/30/2016] [Accepted: 10/12/2016] [Indexed: 12/27/2022]
|
348
|
Riggin CN, Qu F, Kim DH, Huegel J, Steinberg DR, Kuntz AF, Soslowsky LJ, Mauck RL, Bernstein J. Electrospun PLGA Nanofiber Scaffolds Release Ibuprofen Faster and Degrade Slower After In Vivo Implantation. Ann Biomed Eng 2017; 45:2348-2359. [PMID: 28653294 DOI: 10.1007/s10439-017-1876-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/15/2017] [Indexed: 01/04/2023]
Abstract
While delayed delivery of non-steroidal anti-inflammatory drugs (NSAIDs) has been associated with improved tendon healing, early delivery has been associated with impaired healing. Therefore, NSAID use is appropriate only if the dose, timing, and mode of delivery relieves pain but does not impede tissue repair. Because delivery parameters can be controlled using drug-eluting nanofibrous scaffolds, our objective was to develop a scaffold for local controlled release of ibuprofen (IBP), and characterize the release profile and degradation both in vitro and in vivo. We found that when incubated in vitro in saline, scaffolds containing IBP had a linear release profile. However, when implanted subcutaneously in vivo or when incubated in vitro in serum, scaffolds showed a rapid burst release. These data demonstrate that scaffold properties are dependent on the environment in which they are placed and the importance of using serum, rather than saline, for initial in vitro evaluation of biofactor release from biodegradable scaffolds.
Collapse
Affiliation(s)
- Corinne N Riggin
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA.,Department of Bioengineering, University of Pennsylvania, Suite 240 Skirkanich Hall, 210 South 33rd Street, Philadelphia, PA, 19104, USA
| | - Feini Qu
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA.,School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA, 19104, USA
| | - Dong Hwa Kim
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Julianne Huegel
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA
| | - David R Steinberg
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Andrew F Kuntz
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Louis J Soslowsky
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA.,Department of Bioengineering, University of Pennsylvania, Suite 240 Skirkanich Hall, 210 South 33rd Street, Philadelphia, PA, 19104, USA
| | - Robert L Mauck
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA.,Department of Bioengineering, University of Pennsylvania, Suite 240 Skirkanich Hall, 210 South 33rd Street, Philadelphia, PA, 19104, USA
| | - Joseph Bernstein
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA. .,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA.
| |
Collapse
|
349
|
Bohr A, Wang Y, Harmankaya N, Water JJ, Baldursdottír S, Almdal K, Beck-Broichsitter M. Molecular weight-dependent degradation and drug release of surface-eroding poly(ethylene carbonate). Eur J Pharm Biopharm 2017; 115:140-148. [DOI: 10.1016/j.ejpb.2017.02.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Indexed: 02/07/2023]
|
350
|
Sun H, Gu X, Liu K, Fang C, Tang M. Applicability of electrospun polypropylene carbonate polymers as a drug carrier for sirolimus. Mol Med Rep 2017; 15:4253-4258. [PMID: 28487969 DOI: 10.3892/mmr.2017.6540] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 03/08/2017] [Indexed: 11/05/2022] Open
Abstract
Polypropylene carbonate (PPC), a biodegradable aliphatic polyester, exhibits one particular advantage over other polyesters, which is that following degradation in vivo, it primarily produces H2O and CO2, causing minimal side effects. Although PPC exhibits limited mechanical strength, and is therefore not able to serve as a scaffold to support tissue regeneration, it may be suitable for drug delivery; however, this requires further investigation. In the present study, electrospinning was applied to generate PPC polymers containing sirolimus, a cell growth‑inhibiting drug which is used to treat restenosis. The properties of PPC‑sirolimus polymers were examined using scanning electron microscopy, differential scanning calorimetry and in vitro degradation assays. Drug loading and entrapment efficiency were determined, and in vitro sirolimus‑release from the polymer was assessed. Furthermore, the effect of PPC‑sirolimus polymers on cell growth was measured using an MTT assay in vitro. The results of the present study demonstrated that electrospun PPC polymers formed a uniform three‑dimensional, grid‑intertwined, net‑like structure; the surface of the polymers was smooth and the diameter was ~3 µm. Differential scanning calorimetry analysis demonstrated that sirolimus existed in an amorphous state in the polymer. Following soaking in PBS for 4 weeks, the polymer swelled and the net‑like structure broke down and fragmented. Sirolimus loading and entrapment efficiency were 10.3±3.2 and 95.1±10.6%, respectively. Sirolimus‑release from PPC‑sirolimus polymers continued for 28 days in PBS. PPC‑sirolimus markedly inhibited the growth of rat aortic adventitial fibroblast cells, an effect which was not observed with PPC alone. The results of the present study suggest that PPC polymers are a promising alternative drug carrier for sirolimus.
Collapse
Affiliation(s)
- Hourong Sun
- Department of Cardiovascular Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xinghua Gu
- Department of Cardiovascular Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Kai Liu
- Department of Cardiovascular Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Changcun Fang
- Department of Cardiovascular Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Mengmeng Tang
- Department of Cardiovascular Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|