101
|
Ma Y, Sha M, Cheng S, Yao W, Li Z, Qi XR. Construction of Hyaluronic Tetrasaccharide Clusters Modified Polyamidoamine siRNA Delivery System. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E433. [PMID: 29899207 PMCID: PMC6027316 DOI: 10.3390/nano8060433] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 12/25/2022]
Abstract
The CD44 protein, as a predominant receptor for hyaluronan (HA), is highly expressed on the surface of multiple tumor cells. HA, as a targeting molecule for a CD44-contained delivery system, increases intracellular drug concentration in tumor tissue. However, due to the weak binding ability of hyaluronan oligosaccharide to CD44, targeting for tumor drug delivery has been restricted. In this study, we first use a HA tetrasaccharide cluster as the target ligand to enhance the binding ability to CD44. A polyamidoamine (PAMAM) dendrimer was modified by a HA tetrasaccharide cluster as a nonviral vector for small interfering RNA (siRNA) delivery. The dendrimer/siRNA nanocomplexes increased the cellular uptake capacity of siRNA through the CD44 receptor-mediated endocytosis pathway, allowing the siRNA to successfully escape the endosome/lysosome. Compared with the control group, nanocomplexes effectively reduced the expression of GFP protein and mRNA in MDA-MB-231-GFP cells. This delivery system provides a foundation to increase the clinical applications of PAMAM nanomaterials.
Collapse
Affiliation(s)
- Yingcong Ma
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Meng Sha
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Shixuan Cheng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Wang Yao
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Zhongjun Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Xian-Rong Qi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
102
|
Spicer CD, Jumeaux C, Gupta B, Stevens MM. Peptide and protein nanoparticle conjugates: versatile platforms for biomedical applications. Chem Soc Rev 2018; 47:3574-3620. [PMID: 29479622 PMCID: PMC6386136 DOI: 10.1039/c7cs00877e] [Citation(s) in RCA: 283] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Peptide- and protein-nanoparticle conjugates have emerged as powerful tools for biomedical applications, enabling the treatment, diagnosis, and prevention of disease. In this review, we focus on the key roles played by peptides and proteins in improving, controlling, and defining the performance of nanotechnologies. Within this framework, we provide a comprehensive overview of the key sequences and structures utilised to provide biological and physical stability to nano-constructs, direct particles to their target and influence their cellular and tissue distribution, induce and control biological responses, and form polypeptide self-assembled nanoparticles. In doing so, we highlight the great advances made by the field, as well as the challenges still faced in achieving the clinical translation of peptide- and protein-functionalised nano-drug delivery vehicles, imaging species, and active therapeutics.
Collapse
Affiliation(s)
- Christopher D Spicer
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, Stockholm, Sweden.
| | | | | | | |
Collapse
|
103
|
Choi E, Oh J, Lee D, Lee J, Tan X, Kim M, Kim G, Piao C, Lee M. A ternary-complex of a suicide gene, a RAGE-binding peptide, and polyethylenimine as a gene delivery system with anti-tumor and anti-angiogenic dual effects in glioblastoma. J Control Release 2018; 279:40-52. [PMID: 29660374 DOI: 10.1016/j.jconrel.2018.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 03/13/2018] [Accepted: 04/11/2018] [Indexed: 12/20/2022]
Abstract
The receptor for advanced glycation end-products (RAGE) is involved in tumor angiogenesis. Inhibition of RAGE might be an effective anti-angiogenic therapy for cancer. In this study, a cationic RAGE-binding peptide (RBP) was produced as an antagonist of RAGE, and a ternary-complex consisting of RBP, polyethylenimine (2 kDa, PEI2k), and a suicide gene (pHSVtk) was developed as a gene delivery system with dual functions: the anti-tumor effect of pHSVtk and anti-angiogenic effect of RBP. As an antagonist of RAGE, RBP decreased the secretion of vascular-endothelial growth factor (VEGF) in activated macrophages and reduced the tube-formation of endothelial cells in vitro. In in vitro transfection assays, the RBP/PEI2k/plasmid DNA (pDNA) ternary-complex had higher transfection efficiency than the PEI2k/pDNA binary-complex. In an intracranial glioblastoma animal model, the RBP/PEI2k/pHSVtk ternary-complex reduced α-smooth muscle actin expression, suggesting that the complex has an anti-angiogenic effect. In addition, the ternary-complex had higher pHSVtk delivery efficiency than the PEI2k/pHSVtk and PEI25k/pHSVtk binary-complexes in an animal model. As a result, the ternary-complex induced apoptosis and reduced tumor volume more effectively than the PEI2k/pHSVtk and PEI25k/pHSVtk binary-complexes. In conclusion, due to its dual anti-tumor and anti-angiogenesis effects, the RBP/PEI2k/pHSVtk ternary-complex might be an efficient gene delivery system for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Eunji Choi
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Jungju Oh
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Dahee Lee
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Jaewon Lee
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Xiaonan Tan
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Minkyung Kim
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Gyeungyun Kim
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Chunxian Piao
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea.
| |
Collapse
|
104
|
Versatile redox-sensitive pullulan nanoparticles for enhanced liver targeting and efficient cancer therapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1005-1017. [DOI: 10.1016/j.nano.2018.01.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 01/05/2018] [Accepted: 01/15/2018] [Indexed: 11/23/2022]
|
105
|
Wu JY, Wang ZX, Zhang G, Lu X, Qiang GH, Hu W, Ji AL, Wu JH, Jiang CP. Targeted co-delivery of Beclin 1 siRNA and FTY720 to hepatocellular carcinoma by calcium phosphate nanoparticles for enhanced anticancer efficacy. Int J Nanomedicine 2018; 13:1265-1280. [PMID: 29551896 PMCID: PMC5842779 DOI: 10.2147/ijn.s156328] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Purpose FTY720, known as fingolimod, is a new immunosuppressive agent with effective anticancer properties. Although it was recently confirmed that FTY720 inhibits cancer cell proliferation, FTY720 can also induce protective autophagy and reduce cytotoxicity. Blocking autophagy with Beclin 1 siRNA after treatment with FTY720 promotes apoptosis. The objective of this study was to enhance the anticancer effect of FTY720 in hepatocellular carcinoma (HCC) by targeted co-delivery of FTY720 and Beclin 1 siRNA using calcium phosphate (CaP) nanoparticles (NPs). Materials and methods First, the siRNA was encapsulated within the CaP core. To form an asymmetric lipid bilayer structure, we then used an anionic lipid for the inner leaflet and a cationic lipid for the outer leaflet; after removing chloroform by rotary evaporation, these lipids were dispersed in a saline solution with FTY720. The NPs were analyzed by transmission electron microscopy, dynamic light scattering and ultraviolet–visible spectrophotometry. Cancer cell viability and cell death were analyzed by MTT assays, fluorescence-activated cell sorting analysis and Western blotting. In addition, the in vivo effects of the NPs were investigated using an athymic nude mouse subcutaneous transplantation tumor model. Results When the CaP NPs, called LCP-II NPs, were loaded with FTY720 and siRNA, they exhibited the expected size and were internalized by cells. These NPs were stable in systemic circulation. Furthermore, co-delivery of FTY720 and Beclin 1 siRNA significantly increased cytotoxicity in vitro and in vivo compared with that caused by treatment with the free drug alone. Conclusion The CaP NP system can be further developed for co-delivery of FTY720 and Beclin 1 siRNA to treat HCC, enhancing the anticancer efficacy of FTY720. Our findings provide a new insight into HCC treatment with co-delivered small molecules and siRNA, and these results can be readily translated into cancer clinical trials.
Collapse
Affiliation(s)
- Jun-Yi Wu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhong-Xia Wang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Guang Zhang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xian Lu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Guang-Hui Qiang
- Department of Hepatobiliary Surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Hu
- Department of Hepatobiliary Surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - An-Lai Ji
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jun-Hua Wu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Chun-Ping Jiang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
106
|
Oller-Salvia B, Sánchez-Navarro M, Giralt E, Teixidó M. Blood-brain barrier shuttle peptides: an emerging paradigm for brain delivery. Chem Soc Rev 2018; 45:4690-707. [PMID: 27188322 DOI: 10.1039/c6cs00076b] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Brain delivery is one of the major challenges in drug development because of the high number of patients suffering from neural diseases and the low efficiency of the treatments available. Although the blood-brain barrier (BBB) prevents most drugs from reaching their targets, molecular vectors - known as BBB shuttles - offer great promise to safely overcome this formidable obstacle. In recent years, peptide shuttles have received growing attention because of their lower cost, reduced immunogenicity, and higher chemical versatility than traditional Trojan horse antibodies and other proteins.
Collapse
Affiliation(s)
- Benjamí Oller-Salvia
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain.
| | - Macarena Sánchez-Navarro
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain.
| | - Ernest Giralt
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain. and Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain
| | - Meritxell Teixidó
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain.
| |
Collapse
|
107
|
Zheng M, Tao W, Zou Y, Farokhzad OC, Shi B. Nanotechnology-Based Strategies for siRNA Brain Delivery for Disease Therapy. Trends Biotechnol 2018; 36:562-575. [PMID: 29422412 DOI: 10.1016/j.tibtech.2018.01.006] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/10/2018] [Accepted: 01/11/2018] [Indexed: 02/08/2023]
Abstract
Small interfering RNA (siRNA)-based gene silencing technology has demonstrated significant potential for treating brain-associated diseases. However, effective and safe systemic delivery of siRNA into the brain remains challenging because of biological barriers such as enzymatic degradation, short circulation lifetime, the blood-brain barrier (BBB), insufficient tissue penetration, cell endocytosis, and cytosolic transport. Nanotechnology offers intriguing potential for addressing these challenges in siRNA brain delivery in conjunction with chemical and biological modification strategies. In this review, we outline the challenges of systemic delivery of siRNA-based therapy for brain diseases, highlight recent advances in the development and engineering of siRNA nanomedicines for various brain diseases, and discuss our perspectives on this exciting research field for siRNA-based therapy towards more effective brain disease therapy.
Collapse
Affiliation(s)
- Meng Zheng
- International Joint Center for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yan Zou
- International Joint Center for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bingyang Shi
- International Joint Center for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
108
|
Zhang W, Song Y, Eldi P, Guo X, Hayball JD, Garg S, Albrecht H. Targeting prostate cancer cells with hybrid elastin-like polypeptide/liposome nanoparticles. Int J Nanomedicine 2018; 13:293-305. [PMID: 29391790 PMCID: PMC5768422 DOI: 10.2147/ijn.s152485] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Prostate cancer cells frequently overexpress the gastrin-releasing peptide receptor, and various strategies have been applied in preclinical settings to target this receptor for the specific delivery of anticancer compounds. Recently, elastin-like polypeptide (ELP)-based self-assembling micelles with tethered GRP on the surface have been suggested to actively target prostate cancer cells. Poorly soluble chemotherapeutics such as docetaxel (DTX) can be loaded into the hydrophobic cores of ELP micelles, but only limited drug retention times have been achieved. Herein, we report the generation of hybrid ELP/liposome nanoparticles which self-assembled rapidly in response to temperature change, encapsulated DTX at high concentrations with slow release, displayed the GRP ligand on the surface, and specifically bound to GRP receptor expressing PC-3 cells as demonstrated by flow cytometry. This novel type of drug nanocarrier was successfully used to reduce cell viability of prostate cancer cells in vitro through the specific delivery of DTX.
Collapse
Affiliation(s)
- Wei Zhang
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Yunmei Song
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Preethi Eldi
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Xiuli Guo
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - John D Hayball
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia.,Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Sanjay Garg
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Hugo Albrecht
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
109
|
Wang H, Zhong L, Liu Y, Xu X, Xing C, Wang M, Bai SM, Lu CH, Yang HH. A black phosphorus nanosheet-based siRNA delivery system for synergistic photothermal and gene therapy. Chem Commun (Camb) 2018. [DOI: 10.1039/c8cc00931g] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A new siRNA delivery platform based on black phosphorus nanosheets is constructed which shows excellent therapy efficiency for tumors.
Collapse
Affiliation(s)
- Huimeng Wang
- Key Laboratory for Analytical Science of Food Safety and Biology of the MOE
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Lin Zhong
- Key Laboratory for Analytical Science of Food Safety and Biology of the MOE
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Yang Liu
- Key Laboratory for Analytical Science of Food Safety and Biology of the MOE
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Xin Xu
- Key Laboratory for Analytical Science of Food Safety and Biology of the MOE
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Chao Xing
- Key Laboratory for Analytical Science of Food Safety and Biology of the MOE
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Min Wang
- Key Laboratory for Analytical Science of Food Safety and Biology of the MOE
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Shu-Meng Bai
- College of Biological Science and Engineering
- Fuzhou University
- Fuzhou 350116
- P. R. China
| | - Chun-Hua Lu
- Key Laboratory for Analytical Science of Food Safety and Biology of the MOE
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Huang-Hao Yang
- Key Laboratory for Analytical Science of Food Safety and Biology of the MOE
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| |
Collapse
|
110
|
Chun JY, Min SG, Jo YJ. Production of low molecular collagen peptides-loaded liposomes using different charged lipids. Chem Phys Lipids 2017; 209:1-8. [DOI: 10.1016/j.chemphyslip.2017.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 10/06/2017] [Accepted: 10/08/2017] [Indexed: 01/16/2023]
|
111
|
Kim JS, Shin DH, Kim JS. Dual-targeting immunoliposomes using angiopep-2 and CD133 antibody for glioblastoma stem cells. J Control Release 2017; 269:245-257. [PMID: 29162480 DOI: 10.1016/j.jconrel.2017.11.026] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 10/12/2017] [Accepted: 11/17/2017] [Indexed: 01/10/2023]
Abstract
Glioblastoma stem cells (GSCs), which are identified as subpopulation of CD133+/ALDH1+, are known to show resistance to the most of chemotherapy and radiation therapy, leading to the recurrence of tumor in glioblastoma multiforme (GBM) patients. Also, delivery of temozolomide (TMZ), a mainline treatment of GBM, to the GBM site is hampered by various barriers including the blood-brain barrier (BBB). A dual-targeting immunoliposome encapsulating TMZ (Dual-LP-TMZ) was developed by using angiopep-2 (An2) and anti-CD133 monoclonal antibody (CD133 mAb) for BBB transcytosis and specific delivery to GSCs, respectively. The size, zeta potential and drug encapsulation efficiency of Dual-LP-TMZ were 203.4nm in diameter, -1.6mV and 99.2%, respectively. The in vitro cytotoxicity of Dual-LP-TMZ against U87MG GSCs was increased by 425- and 181-folds when compared with that of free TMZ and non-targeted TMZ liposome (LP-TMZ) (10.3μM vs. 4380μM and 1869μM in IC50, respectively). Apoptosis and anti-migration ability of Dual-LP-TMZ in U87MG GSCs were also significantly enhanced comparing with those of free TMZ or LP-TMZ. In vivo study clearly showed a significant reduction in tumor size after intravenous administrations of Dual-LP-TMZ to the orthotopically-implanted brain tumor mice when compared with free TMZ or LP-TMZ. Increased life span (ILS) and median survival time (MST) of tumor-bearing mice were also increased when treated with Dual-LP-TMZ (211.2% in ILS and 49.2days in MST) than with free TMZ (0% in ILS and 23.3day in MST). These data indicate that conjugation of both An2 peptide and CD133 mAb to TMZ-encapsulating liposome is very effective in delivering the TMZ to GSCs via BBB, suggesting a potential use of Dual-LP-TMZ as a therapeutic modality for GBM.
Collapse
Affiliation(s)
- Jung Seok Kim
- Research Center for Cell Fate Control (RCCFC), Drug Information Research Institute (DIRI), College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Dae Hwan Shin
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Jin-Seok Kim
- Research Center for Cell Fate Control (RCCFC), Drug Information Research Institute (DIRI), College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea.
| |
Collapse
|
112
|
Chan L, He L, Zhou B, Guan S, Bo M, Yang Y, Liu Y, Liu X, Zhang Y, Xie Q, Chen T. Cancer-Targeted Selenium Nanoparticles Sensitize Cancer Cells to Continuous γ Radiation to Achieve Synergetic Chemo-Radiotherapy. Chem Asian J 2017; 12:3053-3060. [PMID: 28892302 DOI: 10.1002/asia.201701227] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/09/2017] [Indexed: 12/23/2022]
Abstract
Cancer radiotherapy with 125 I seeds demonstrates higher long-term efficacy and fewer side effects than traditional X-ray radiotherapy owing to its low-dose and continuous radiation but is still limited by radioresistance in clinical applications. Therefore, the design and synthesis of sensitizers that could enhance the sensitivity of cancer cells to 125 I seeds is of great importance for future radiotherapy. Selenium nanoparticles (SeNPs) have been found to exhibit high potential in cancer chemotherapy and as drug carriers. In this study, we found that, based on the Auger-electron effect and Compton effect of Se atoms, cancer-targeted SeNPs in combination with 125 I seeds achieve synergetic effects to inhibit cancer-cell growth and colony formation through the induction of cell apoptosis and cell cycle arrest. Detailed studies on the action mechanisms reveal that the combined treatments effectively activate intracellular reactive oxygen species (ROS) overproduction to regulate p53-mediated DNA damage apoptotic signaling pathways and mitogen-activated protein kinase (MAPK) phosphorylation and to prevent the self-repair of cancer cells simultaneously. Taken together, the combination of SeNPs with 125 I seeds could be further exploited as a safe and effective strategy for next-generation cancer chemo-radiotherapy in clinical applications.
Collapse
Affiliation(s)
- Leung Chan
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Lizhen He
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Binwei Zhou
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Shouhai Guan
- The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Mingjun Bo
- The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yahui Yang
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Ying Liu
- Wu Jing Zong Dui Hospital of Guangdong Province, Guangzhou, 510507, China
| | - Xiao Liu
- Wu Jing Zong Dui Hospital of Guangdong Province, Guangzhou, 510507, China
| | - Yanyang Zhang
- The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Qiang Xie
- The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
113
|
Multifunctional nanoparticles co-delivering EZH2 siRNA and etoposide for synergistic therapy of orthotopic non-small-cell lung tumor. J Control Release 2017; 268:198-211. [PMID: 29061511 DOI: 10.1016/j.jconrel.2017.10.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 05/17/2017] [Accepted: 10/14/2017] [Indexed: 12/29/2022]
Abstract
Malignant proliferation and metastasis in non-small cell lung carcinoma (NSCLC) are great challenges for effective clinical treatment through conventional chemotherapy. The combinational therapy strategy of RNA interfering (RNAi) technology and chemotherapeutic agents have been reported to be promising for effective cancer therapy. In this study, based on multifunctional nanoparticles (NPs), the simultaneous delivery of etoposide (ETP) and anti-Enhancer of Zeste Homologue 2 (EZH2) siRNA for the effective treatment of orthotopic lung tumor was achieved. The NPs exhibited pH/redox dual sensitivity verified by particle size changes, morphological changes, and in vitro release of drugs. Confocal microscopy analysis confirmed that the NPs exhibited endosomal escape property and on-demand intracellular drug release behavior, which can protect siRNA from degradation and facilitate the chemotherapeutic effect respectively. In vitro tumor cell motility study demonstrated that EZH2 siRNA loaded in NPs can decrease the migration and invasion capabilities of tumor cells by downregulating the expression of EZH2 mRNA and protein. In particular, an antiproliferation study revealed that the co-delivery of siRNA and ETP in the multifunctional NPs can induce a synergistic therapeutic effect on NSCLC. In vivo targeting evaluation showed that cRGDyC-PEG modification on NPs exhibited a low distribution in normal organs and an obvious accumulation in orthotopic lung tumor. Furthermore, targeted NPs co-delivering siRNA and ETP showed superior inhibition on tumor growth and metastasis and produced minimal systemic toxicity. These findings indicated that multifunctional NPs can be utilized as a co-delivery system, and that the combination of EZH2 siRNA and ETP can effectively treat NSCLC.
Collapse
|
114
|
Kydd J, Jadia R, Velpurisiva P, Gad A, Paliwal S, Rai P. Targeting Strategies for the Combination Treatment of Cancer Using Drug Delivery Systems. Pharmaceutics 2017; 9:E46. [PMID: 29036899 PMCID: PMC5750652 DOI: 10.3390/pharmaceutics9040046] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/01/2017] [Accepted: 10/10/2017] [Indexed: 12/20/2022] Open
Abstract
Cancer cells have characteristics of acquired and intrinsic resistances to chemotherapy treatment-due to the hostile tumor microenvironment-that create a significant challenge for effective therapeutic regimens. Multidrug resistance, collateral toxicity to normal cells, and detrimental systemic side effects present significant obstacles, necessitating alternative and safer treatment strategies. Traditional administration of chemotherapeutics has demonstrated minimal success due to the non-specificity of action, uptake and rapid clearance by the immune system, and subsequent metabolic alteration and poor tumor penetration. Nanomedicine can provide a more effective approach to targeting cancer by focusing on the vascular, tissue, and cellular characteristics that are unique to solid tumors. Targeted methods of treatment using nanoparticles can decrease the likelihood of resistant clonal populations of cancerous cells. Dual encapsulation of chemotherapeutic drug allows simultaneous targeting of more than one characteristic of the tumor. Several first-generation, non-targeted nanomedicines have received clinical approval starting with Doxil® in 1995. However, more than two decades later, second-generation or targeted nanomedicines have yet to be approved for treatment despite promising results in pre-clinical studies. This review highlights recent studies using targeted nanoparticles for cancer treatment focusing on approaches that target either the tumor vasculature (referred to as 'vascular targeting'), the tumor microenvironment ('tissue targeting') or the individual cancer cells ('cellular targeting'). Recent studies combining these different targeting methods are also discussed in this review. Finally, this review summarizes some of the reasons for the lack of clinical success in the field of targeted nanomedicines.
Collapse
Affiliation(s)
- Janel Kydd
- Department of Biomedical Engineering and Biotechnology, University of Massachusetts, 1 University Ave, Lowell, MA 01854, USA.
| | - Rahul Jadia
- Department of Biomedical Engineering and Biotechnology, University of Massachusetts, 1 University Ave, Lowell, MA 01854, USA.
| | - Praveena Velpurisiva
- Department of Biomedical Engineering and Biotechnology, University of Massachusetts, 1 University Ave, Lowell, MA 01854, USA.
| | - Aniket Gad
- Confocal Imaging Core, Beth Israel Deaconess Medical Center, 330 Brookline Avenue Boston, MA 02215, USA.
| | - Shailee Paliwal
- Department of Chemical Engineering, University of Massachusetts, 1 University Ave, Lowell, MA 01854, USA.
| | - Prakash Rai
- Department of Biomedical Engineering and Biotechnology, University of Massachusetts, 1 University Ave, Lowell, MA 01854, USA.
- Department of Chemical Engineering, University of Massachusetts, 1 University Ave, Lowell, MA 01854, USA.
| |
Collapse
|
115
|
Játiva P, Ceña V. Use of nanoparticles for glioblastoma treatment: a new approach. Nanomedicine (Lond) 2017; 12:2533-2554. [DOI: 10.2217/nnm-2017-0223] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is a very aggressive CNS tumor with poor prognosis. Current treatment lacks efficacy indicating that new therapeutic approaches are needed. One of these new approaches is based on the use of nanoparticles (NPs) to deliver different cargos (antitumoral drugs or genetic materials) to tumoral cells. This review covers the signaling pathways altered in GBM cells to understand the rationale behind choosing new therapeutic targets and recent advances in the use of different NPs to deliver to GBM cells, both in vitro and in vivo, different therapeutic molecules. A special focus is placed on the effect of NPs on orthotopic brain tumors since this animal model represents the optimal model for translational purposes.
Collapse
Affiliation(s)
- Pablo Játiva
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, Albacete, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Valentín Ceña
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, Albacete, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
116
|
Babu A, Munshi A, Ramesh R. Combinatorial therapeutic approaches with RNAi and anticancer drugs using nanodrug delivery systems. Drug Dev Ind Pharm 2017; 43:1391-1401. [PMID: 28523942 PMCID: PMC6101010 DOI: 10.1080/03639045.2017.1313861] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 12/31/2022]
Abstract
RNA interference (RNAi) is emerging as a powerful approach in cancer treatment. siRNA is an important RNAi tool that can be designed to specifically silence the expression of genes involved in drug resistance and chemotherapeutic inactivity. Combining siRNA and other therapeutic agents can overcome the multidrug resistance (MDR) phenomenon by simultaneously silencing genes and enhancing chemotherapeutic activity. Moreover, the therapeutic efficiency of anticancer drugs can be significantly improved by additive or synergistic effects induced by siRNA and combined therapies. Co-delivery of these diverse anticancer agents, however, requires specially designed nanocarriers. This review highlights the recent trends in siRNA/anticancer drug co-delivery systems under the major categories of liposomes/lipid, polymeric and inorganic nanoplatforms. The objective is to discuss the strategies for nanocarrier-based co-delivery systems using siRNA/anticancer drug combinations, emphasizing various siRNA targets that help overcome MDR and enhance therapeutic efficiency.
Collapse
Affiliation(s)
- Anish Babu
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
- Department of Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
| | - Anupama Munshi
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
- Department of Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
| | - Rajagopal Ramesh
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
- Department of Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
- Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
| |
Collapse
|
117
|
Wang M, Wang J, Li B, Meng L, Tian Z. Recent advances in mechanism-based chemotherapy drug-siRNA pairs in co-delivery systems for cancer: A review. Colloids Surf B Biointerfaces 2017; 157:297-308. [DOI: 10.1016/j.colsurfb.2017.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022]
|
118
|
Liang DS, Zhang WJ, Wang AT, Su HT, Zhong HJ, Qi XR. Treating metastatic triple negative breast cancer with CD44/neuropilin dual molecular targets of multifunctional nanoparticles. Biomaterials 2017; 137:23-36. [DOI: 10.1016/j.biomaterials.2017.05.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 05/12/2017] [Accepted: 05/12/2017] [Indexed: 12/18/2022]
|
119
|
Huang L, Hu J, Huang S, Wang B, Siaw-Debrah F, Nyanzu M, Zhang Y, Zhuge Q. Nanomaterial applications for neurological diseases and central nervous system injury. Prog Neurobiol 2017; 157:29-48. [PMID: 28743465 DOI: 10.1016/j.pneurobio.2017.07.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/20/2022]
Abstract
The effectiveness of noninvasive treatment for neurological disease is generally limited by the poor entry of therapeutic agents into the central nervous system (CNS). Most CNS drugs cannot permeate into the brain parenchyma because of the blood-brain barrier thus, overcoming this problem has become one of the most significant challenges in the development of neurological therapeutics. Nanotechnology has emerged as an innovative alternative for treating neurological diseases. In fact, rapid advances in nanotechnology have provided promising solutions to this challenge. This review highlights the applications of nanomaterials in the developing neurological field and discusses the evidence for their efficacies.
Collapse
Affiliation(s)
- Lijie Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Jiangnan Hu
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Shengwei Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Brian Wang
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Felix Siaw-Debrah
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Mark Nyanzu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Yu Zhang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Qichuan Zhuge
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China.
| |
Collapse
|
120
|
Han W, Yin G, Pu X, Chen X, Liao X, Huang Z. Glioma targeted delivery strategy of doxorubicin-loaded liposomes by dual-ligand modification. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:1695-1712. [PMID: 28699828 DOI: 10.1080/09205063.2017.1348739] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The blood-brain barrier (BBB) is the protective parclose of brain safety, but it is also the main obstacle of the drug delivery to cerebral parenchyma, which hamper therapy for brain diseases. In this work, a glioma targeted drug delivery system was developed through loading doxorubicin into Angiopep-2 and TAT peptide dual-modified liposomes (DOX-TAT-Ang-LIP). Low-density lipoprotein receptor-related protein-1 (LRP1) was one receptor overexpressed on both BBB and glioma cytomembranes. Angiopep-2, a specific ligand of LRP1, exhibited high LRP1 binding efficiency. Additionally, TAT could penetrate through cell membranes without selectivity via an unsaturated pathway. To avoid the receptor saturation of Angiopep-2, TAT was also conjugated on the surface of liposomes, providing that the liposomes not only have effective BBB penetrating effect, but also have the glioma targeting function. The prepared DOX liposomes appeared good stability and narrow dispersity in serum with a diameter of 90 nm, and exhibited sustained DOX release behaviors. The conjunctions of Angiopep-2 and TAT were confirmed by 1H NMR spectra. The BBB model, cellular uptake observations, antiproliferation study, and the cell ultrastructure analyses suggested that DOX-TAT-Ang-LIP could not only penetrate through BBB via transcytosis, but also concentrate in glioma, then enter into glioma cells and finally result in the necrosis of glioma cells.
Collapse
Affiliation(s)
- Wei Han
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| | - Guangfu Yin
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| | - Ximing Pu
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| | - Xianchun Chen
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| | - Xiaoming Liao
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| | - Zhongbing Huang
- a College of Materials Science and Engineering, Sichuan University , Chengdu , China
| |
Collapse
|
121
|
Chen X, Zhang Y, Tang C, Tian C, Sun Q, Su Z, Xue L, Yin Y, Ju C, Zhang C. Co-delivery of paclitaxel and anti-survivin siRNA via redox-sensitive oligopeptide liposomes for the synergistic treatment of breast cancer and metastasis. Int J Pharm 2017. [PMID: 28642204 DOI: 10.1016/j.ijpharm.2017.06.071] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The overexpression of survivin in breast cancer cells is an important factor of paclitaxel (PTX) resistance in breast cancer. To overcome PTX resistance and improve the antitumor effect of PTX, we developed a novel liposome-based nanosystem (PTX/siRNA/SS-L), composed of a redox-sensitive cationic oligopeptide lipid (LHSSG2C14) with a proton sponge effect, natural soybean phosphatidylcholine (SPC), and cholesterol for co-delivery of PTX and anti-survivin siRNA, which could specifically downregulate survivin overexpression. PTX/siRNA/SS-L exhibited high encapsulation efficiency and rapid redox-responsive release of both PTX and siRNA. Moreover, in vitro studies on the 4T1 breast cancer cells revealed that PTX/siRNA/SS-L offered significant advantages over other experimental groups, such as higher cellular uptake, successful endolysosomal escape, reduced survivin expression, the lowest cell viability and wound healing rate, as well as the highest apoptosis rate. In particular, in vivo evaluation of 4T1 tumor-bearing mice showed that PTX/siRNA/SS-L had lower toxicity and induced a synergistic inhibitory effect on tumor growth and pulmonary metastasis. Collectively, the collaboration of anti-survivin siRNA and PTX via redox-sensitive oligopeptide liposomes provides a promising strategy for the treatment of breast cancer and metastasis.
Collapse
Affiliation(s)
- Xinyan Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China; Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yidi Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Chunming Tang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Chunli Tian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Qiong Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Zhigui Su
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Yifan Yin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Caoyun Ju
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China.
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210046, China.
| |
Collapse
|
122
|
Photoluminescent and biodegradable polycitrate-polyethylene glycol-polyethyleneimine polymers as highly biocompatible and efficient vectors for bioimaging-guided siRNA and miRNA delivery. Acta Biomater 2017; 54:69-80. [PMID: 28219808 DOI: 10.1016/j.actbio.2017.02.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/15/2017] [Accepted: 02/16/2017] [Indexed: 11/23/2022]
Abstract
Development of biodegradable and biocompatible non-viral vectors with intrinsical multifunctional properties such as bioimaging ability for highly efficient nucleic acids delivery still remains a challenge. Here, a biodegradable poly (1,8-octanedio-citric acid)-co-polyethylene glycol grafted with polyethyleneimine (PEI) (POCG-PEI) polymers with the photoluminescent capacity were synthesized for nucleic acids delivery (siRNA and miRNA). POCG-PEI polymers can efficiently bind various nucleic acids, protect them against enzymatic degradation and release the genes in the presence of polyanionic heparin. POCG-PEI also showed a significantly low cytotoxicity, enhanced cellular uptake and high transfection efficiency of nucleic acids, as compared to commercial transfection agents, lipofectamine 2000 (Lipo) and polyethylenimine (PEI 25K). POCG-PEI polymers demonstrate an excellent photostability, which allows for imaging the cells and real-time tracking the nucleic acids delivery. The photoluminescent property, low cytotoxicity, biodegradation, good gene binding and protection ability and high genes delivery efficiency make POCG-PEI highly competitive as a non-virus vector for genes delivery and real-time bioimaging applications. Our results may be also an important step for designing biodegradable biomaterials with multifunctional properties towards bioimaging-guided genes therapeutic applications. STATEMENT OF SIGNIFICANCE Here, a biodegradable poly (1,8-octanedio-citric acid)-co-polyethylene glycol grafted with polyethyleneimine (PEI) (POCG-PEI) polymers with controlled photoluminescent capacity were synthesized for nucleic acids delivery (siRNA and miRNA). POCG-PEI polymers can efficiently bind various nucleic acids, protect them against enzymatic degradation and release the genes in the presence of polyanionic heparin. POCG-PEI also showed a significantly low cytotoxicity, enhanced cellular uptake and high transfection efficiency of nucleic acids, as compared to commercial transfection agents, lipofectamine 2000 (Lipo) and polyethylenimine (PEI 25K). POCG-PEI polymers demonstrate an excellent photostability, which allows for imaging the cells and real-time tracking the nucleic acids delivery. Our results may be also an important step for designing biodegradable biomaterials with multifunctional properties towards bioimaging-guided genes therapeutic applications.
Collapse
|
123
|
Surface modification of lipid-based nanocarriers for cancer cell-specific drug targeting. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0329-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
124
|
Brain-Targeted Polymers for Gene Delivery in the Treatment of Brain Diseases. Top Curr Chem (Cham) 2017; 375:48. [PMID: 28397188 DOI: 10.1007/s41061-017-0138-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/27/2017] [Indexed: 10/19/2022]
Abstract
Gene therapies have become a promising strategy for treating neurological disorders, such as brain cancer and neurodegenerative diseases, with the help of molecular biology interpreting the underlying pathological mechanisms. Successful cellular manipulation against these diseases requires efficient delivery of nucleic acids into brain and further into specific neurons or cancer cells. Compared with viral vectors, non-viral polymeric carriers provide a safer and more flexible way of gene delivery, although suffering from significantly lower transfection efficiency. Researchers have been devoted to solving this defect, which is attributed to the multiple barriers existing for gene therapeutics in vivo, such as systemic degradation, blood-brain barrier, and endosome trapping. This review will be mainly focused on systemically administrated brain-targeted polymers developed so far, including PEI, dendrimers, and synthetic polymers with various functions. We will discuss in detail how they are designed to overcome these barriers and how they efficiently deliver therapeutic nucleic acids into targeted cells.
Collapse
|
125
|
Wu B, Lu ST, Zhang LJ, Zhuo RX, Xu HB, Huang SW. Codelivery of doxorubicin and triptolide with reduction-sensitive lipid-polymer hybrid nanoparticles for in vitro and in vivo synergistic cancer treatment. Int J Nanomedicine 2017; 12:1853-1862. [PMID: 28331310 PMCID: PMC5352248 DOI: 10.2147/ijn.s131235] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Codelivery is a promising strategy to overcome the limitations of single chemotherapeutic agents in cancer treatment. Despite progress, codelivery of two or more different functional drugs to increase anticancer efficiency still remains a challenge. Here, reduction-sensitive lipid–polymer hybrid nanoparticles (LPNPs) drug delivery system composed of monomethoxy-poly(ethylene glycol)-S-S-hexadecyl (mPEG-S-S-C16), soybean lecithin, and poly(D,L-lactide-co-glycolide) (PLGA) was used for codelivery of doxorubicin (DOX) and a Chinese herb extract triptolide (TPL). Hydrophobic DOX and TPL could be successfully loaded in LPNPs by self-assembly. More importantly, drug release and cellular uptake experiments demonstrated that the two drugs were reduction sensitive, released simultaneously from LPNPs, and taken up effectively by the tumor cells. DOX/TPL-coloaded LPNPs (DOX/TPL-LPNPs) exhibited a high level of synergistic activation with low combination index (CI) in vitro and in vivo. Moreover, the highest synergistic therapeutic effect was achieved at the ratio of 1:0.2 DOX/TPL. Further experiments showed that TPL enhanced the uptake of DOX by human oral cavity squamous cell carcinoma cells (KB cells). Overall, DOX/TPL-coencapsulated reduction-sensitive nanoparticles will be a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Bo Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University; Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, People's Republic of China
| | - Shu-Ting Lu
- Department of Radiology, Zhongnan Hospital of Wuhan University
| | - Liu-Jie Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, People's Republic of China
| | - Ren-Xi Zhuo
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, People's Republic of China
| | - Hai-Bo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University
| | - Shi-Wen Huang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
126
|
Yang ZZ, Gao W, Liu YJ, Pang N, Qi XR. Delivering siRNA and Chemotherapeutic Molecules Across BBB and BTB for Intracranial Glioblastoma Therapy. Mol Pharm 2017; 14:1012-1022. [DOI: 10.1021/acs.molpharmaceut.6b00819] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Zhen-zhen Yang
- Beijing
Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System,
Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wei Gao
- Beijing
Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System,
Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yu-jie Liu
- Beijing
Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System,
Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ning Pang
- Beijing
Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System,
Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xian-rong Qi
- Beijing
Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System,
Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| |
Collapse
|
127
|
Bredlau AL, Dixit S, Chen C, Broome AM. Nanotechnology Applications for Diffuse Intrinsic Pontine Glioma. Curr Neuropharmacol 2017; 15:104-115. [PMID: 26903150 PMCID: PMC5327462 DOI: 10.2174/1570159x14666160223121002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 10/12/2015] [Accepted: 01/30/2016] [Indexed: 12/19/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are invariably fatal tumors found in the pons of elementary school aged children. These tumors are grade II-IV gliomas, with a median survival of less than 1 year from diagnosis when treated with standard of care (SOC) therapy. Nanotechnology may offer therapeutic options for the treatment of DIPGs. Multiple nanoparticle formulations are currently being investigated for the treatment of DIPGs. Nanoparticles based upon stable elements, polymer nanoparticles, and organic nanoparticles are under development for the treatment of brain tumors, including DIPGs. Targeting of nanoparticles is now possible as delivery techniques that address the difficulty in crossing the blood brain barrier (BBB) are developed. Theranostic nanoparticles, a combination of therapeutics and diagnostic nanoparticles, improve imaging of the cancerous tissue while delivering therapy to the local region. However, additional time and attention should be directed to developing a nanoparticle delivery system for treatment of the uniformly fatal pediatric disease of DIPG.
Collapse
Affiliation(s)
| | | | | | - Ann-Marie Broome
- Department of Radiology and Radiological Sciences, Medical University of South Carolina, 68 President Street, MSC 120/BEB 213, Charleston, SC 29425, USA
| |
Collapse
|
128
|
Kuang H, Ku SH, Kokkoli E. The design of peptide-amphiphiles as functional ligands for liposomal anticancer drug and gene delivery. Adv Drug Deliv Rev 2017; 110-111:80-101. [PMID: 27539561 DOI: 10.1016/j.addr.2016.08.005] [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: 05/02/2016] [Revised: 07/12/2016] [Accepted: 08/05/2016] [Indexed: 12/25/2022]
Abstract
Liposomal nanomedicine has led to clinically useful cancer therapeutics like Doxil and DaunoXome. In addition, peptide-functionalized liposomes represent an effective drug and gene delivery vehicle with increased cancer cell specificity, enhanced tumor-penetrating ability and high tumor growth inhibition. The goal of this article is to review the recently published literature of the peptide-amphiphiles that were used to functionalize liposomes, to highlight successful designs that improved drug and gene delivery to cancer cells in vitro, and cancer tumors in vivo, and to discuss the current challenges of designing these peptide-decorated liposomes for effective cancer treatment.
Collapse
|
129
|
Ma Y, Liu Y, Sun A, Du Y, Ye M, Pu X, Qi X. Intestinal absorption and neuroprotective effects of kaempferol-3-O-rutinoside. RSC Adv 2017. [DOI: 10.1039/c7ra05415g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Kaempferol-3-O-rutinoside (K3R) has been proven to have biological activities for the prevention and treatment of central nervous system (CNS) diseases.
Collapse
Affiliation(s)
- Yingcong Ma
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System
- Department of Pharmaceutics
- School of Pharmaceutical Sciences
- Peking University
- Beijing
| | - Yujie Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System
- Department of Pharmaceutics
- School of Pharmaceutical Sciences
- Peking University
- Beijing
| | - Aning Sun
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System
- Department of Pharmaceutics
- School of Pharmaceutical Sciences
- Peking University
- Beijing
| | - Yitian Du
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System
- Department of Pharmaceutics
- School of Pharmaceutical Sciences
- Peking University
- Beijing
| | - Min Ye
- Department of Natural Medicines
- School of Pharmaceutical Sciences
- Peking University
- Beijing
- PR China
| | - Xiaoping Pu
- Department of Molecular and Cellular Pharmacology
- School of Pharmaceutical Sciences
- Peking University
- Beijing
- PR China
| | - Xianrong Qi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System
- Department of Pharmaceutics
- School of Pharmaceutical Sciences
- Peking University
- Beijing
| |
Collapse
|
130
|
Wang D, Wu LP. Nanomaterials for delivery of nucleic acid to the central nervous system (CNS). MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 70:1039-1046. [DOI: 10.1016/j.msec.2016.04.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/11/2016] [Accepted: 04/04/2016] [Indexed: 11/08/2022]
|
131
|
He C, Cai P, Li J, Zhang T, Lin L, Abbasi AZ, Henderson JT, Rauth AM, Wu XY. Blood-brain barrier-penetrating amphiphilic polymer nanoparticles deliver docetaxel for the treatment of brain metastases of triple negative breast cancer. J Control Release 2017; 246:98-109. [DOI: 10.1016/j.jconrel.2016.12.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 12/19/2016] [Indexed: 12/29/2022]
|
132
|
Sahu AK, Jain V. Screening of process variables using Plackett-Burman design in the fabrication of gedunin-loaded liposomes. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:1011-1022. [PMID: 27917681 DOI: 10.1080/21691401.2016.1200057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
This study is to screening the formulation and process variables that produce significant effect on the gedunin-loaded liposome formulations by using quality-by-design approach. Placket-Burman screening design was used to screen the most influencing formulation and process variables. Mean vesicle size, zeta potential, entrapment efficiency, and loading capacity were found in the range of 112-990 nm, -19.39 to -39.20 mV, 45.25-87.60%, and 3.54-10.47%, respectively. Differential scanning calorimetry (DSC) and powder X-ray diffraction (XRD) result suggested that Gedunin encapsulated within liposome as amorphous state. The analysis of Pareto chart represented that selected independent variables had a most significant effect on dependent variables.
Collapse
Affiliation(s)
- Anil Kumar Sahu
- a Department of Pharmacy , University Institute of Pharmacy, Affiliated by Pt. Ravishankar Shukla University , Raipur, Chhattisgarh , India
| | - Vishal Jain
- a Department of Pharmacy , University Institute of Pharmacy, Affiliated by Pt. Ravishankar Shukla University , Raipur, Chhattisgarh , India
| |
Collapse
|
133
|
Yin T, Liu J, Zhao Z, Dong L, Cai H, Yin L, Zhou J, Huo M. Smart nanoparticles with a detachable outer shell for maximized synergistic antitumor efficacy of therapeutics with varying physicochemical properties. J Control Release 2016; 243:54-68. [DOI: 10.1016/j.jconrel.2016.09.036] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/29/2016] [Accepted: 09/30/2016] [Indexed: 01/07/2023]
|
134
|
A DNA dual lock-and-key strategy for cell-subtype-specific siRNA delivery. Nat Commun 2016; 7:13580. [PMID: 27882923 PMCID: PMC5476801 DOI: 10.1038/ncomms13580] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 10/17/2016] [Indexed: 02/07/2023] Open
Abstract
The efficient and precise delivery of siRNA to target cells is critical to successful gene therapy. While novel nanomaterials enhance delivery efficiency, it still remains challenging for precise gene delivery to overcome nonspecific adsorption and off-target effect. Here we design a dual lock-and-key system to perform cell-subtype-specific recognition and siRNA delivery. The siRNA is self-assembled in an oligonucleotide nano vehicle that is modified with a hairpin structure to act as both the ‘smart key’ and the delivery carrier. The auto-cleavable hairpin structure can be activated on site at target cell membrane by reacting with two aptamers as ‘dual locks’ sequentially, which leads to cell-subtype discrimination and precise siRNA delivery for high efficient gene silencing. The success of this strategy demonstrates the precise delivery of siRNA to specific target cells by controlling multiple parameters, thus paving the way for application of RNAi in accurate diagnosis and intervention. Delivery of siRNA to target cells is essential for in vivo gene therapy. Here the authors demonstrate an oligonucleotide aptamer that targets therapeutic siRNA to a specific cell type.
Collapse
|
135
|
Alibolandi M, Abnous K, Hadizadeh F, Taghdisi SM, Alabdollah F, Mohammadi M, Nassirli H, Ramezani M. Dextran-poly lactide- co -glycolide polymersomes decorated with folate-antennae for targeted delivery of docetaxel to breast adenocarcinima in vitro and in vivo. J Control Release 2016; 241:45-56. [DOI: 10.1016/j.jconrel.2016.09.012] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/04/2016] [Accepted: 09/12/2016] [Indexed: 11/16/2022]
|
136
|
Wanjale MV, Kumar GSV. Peptides as a therapeutic avenue for nanocarrier-aided targeting of glioma. Expert Opin Drug Deliv 2016; 14:811-824. [PMID: 27690671 DOI: 10.1080/17425247.2017.1242574] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Very few successful interventions have been possible in glioma therapy owing to its aggressive nature as well as its hindrance of targeted therapy together with the limited access afforded by the blood-brain barrier (BBB). With the advent of nanotechnology based delivery vehicles such as micelles, dendrimers, polymer-based nanoparticles and nanogels, the breach of the BBB has been facilitated. However, there remains the issue of targeted therapy for glioma cells. Peptide-mediated surface modification of nanocarriers serves this purpose, extending the ability to target glioma further than the enhanced permeability and retention effect. Areas covered: Here we have tried to re-establish the significance of peptides that could be used in various ways for treating glioma. Peptide-embellished nanocarriers used to deliver anticancer drugs; nucleic acids (siRNA, miRNA); micelles or dendrimers grafted with immunogenic glioma-derived peptides used for stimulating active immunity in vaccine therapy, glioma targets for cell penetrating peptides and homing to specific receptors are reviewed. Expert opinion: Peptides have multifunctional potential in targeting, BBB and cell penetration, and can serve as antagonists of various ligands and agonists of particular over-expressed receptors as discussed in this review. Using peptides in targeted personalized therapy would be one step forward and may offer new avenues for glioma therapeutics.
Collapse
Affiliation(s)
- Mrunal Vitthal Wanjale
- a Chemical Biology, Nano Drug Delivery Systems, Bio-Innovation Center (BIC) , Rajiv Gandhi Centre for Biotechnology , Thiruvananthapuram , Kerala , India
| | - G S Vinod Kumar
- a Chemical Biology, Nano Drug Delivery Systems, Bio-Innovation Center (BIC) , Rajiv Gandhi Centre for Biotechnology , Thiruvananthapuram , Kerala , India
| |
Collapse
|
137
|
Li J, Cheng X, Chen Y, He W, Ni L, Xiong P, Wei M. Vitamin E TPGS modified liposomes enhance cellular uptake and targeted delivery of luteolin: An in vivo/in vitro evaluation. Int J Pharm 2016; 512:262-272. [DOI: 10.1016/j.ijpharm.2016.08.037] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 08/01/2016] [Accepted: 08/17/2016] [Indexed: 01/12/2023]
|
138
|
Xu HL, Mao KL, Lu CT, Fan ZL, Yang JJ, Xu J, Chen PP, ZhuGe DL, Shen BX, Jin BH, Xiao J, Zhao YZ. An injectable acellular matrix scaffold with absorbable permeable nanoparticles improves the therapeutic effects of docetaxel on glioblastoma. Biomaterials 2016; 107:44-60. [PMID: 27614158 DOI: 10.1016/j.biomaterials.2016.08.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/13/2016] [Accepted: 08/16/2016] [Indexed: 10/21/2022]
Abstract
Intratumoral drug delivery (IT) is an inherently appealing approach for concentrating toxic chemotherapies at the site of action. However, for most chemotherapies, poor tumor penetration and short retention at the administration site limit their anti-tumor effects. In this work, we describe permeable nanoparticles (NPs) prepared with a novel amphiphilic polymer, RRR-α-tocopheryl succinate-grafted-ε-polylysine conjugate (VES-g-ε-PLL). The nanoparticles (NPs) of VES-g-ε-PLL exhibited an ultra-small hydrodynamic diameter (20.8 nm) and positive zeta potential (20.6 mV), which facilitate strong glioma spheroid penetration ability in vitro. Additionally, the hydrophobic model drug docetaxel (DTX) could be effectively encapsulated in the nanoparticles with 3.99% drug loading and 73.37% encapsulation efficiency. To prolong the retention time of DTX-loaded nanoparticles (DTX-NPs) in the tumor, intact decellularized brain extracellular matrix (dBECM) derived from healthy rats was used as a drug depot to adsorb the ultra-small DTX-NPs. The intact DTX-NPs-adsorbing dBECM scaffold was further homogenized into an injectable DTX-NPs-dBECM suspension for intratumoral administration. The DTX-NPs-dBECM suspension exhibited slower DTX release than naked DTX-NPs without compromising the tumor penetration ability of DTX-NPs. An antitumor study showed that the DTX-NPs-dBECM suspension exhibited more powerful in vitro inhibition of tumor spheroid growth than free DTX solution or DTX-NPs. Due to strong tumor penetration ability and prolonged retention, DTX-NPs-dBECM led to complete suppression of glioma growth in vivo at 28 days after treatment. The therapeutic mechanism was due to enhanced proliferation inhibition and apoptosis of tumor cells and angiogenesis inhibition of glioma after treatment with DTX-NPs-dBECM. Finally, the safety of DTX-NPs-dBECM at the therapeutic dose was demonstrated via pathological HE assay from heart, liver, spleen, lung and kidney tissues. In conclusion, permeable nanoparticle-absorbing dBECM is a potential carrier for intratumoral delivery of common chemotherapeutics.
Collapse
Affiliation(s)
- He-Lin Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Kai-Li Mao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Cui-Tao Lu
- The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China.
| | - Zi-Liang Fan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Jing-Jing Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Jie Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Pian-Pian Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - De-Li ZhuGe
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Bi-Xin Shen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Bing-Hui Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China.
| | - Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China; The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China.
| |
Collapse
|
139
|
Meng F, Han N, Yeo Y. Organic nanoparticle systems for spatiotemporal control of multimodal chemotherapy. Expert Opin Drug Deliv 2016; 14:427-446. [PMID: 27476442 DOI: 10.1080/17425247.2016.1218464] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Chemotherapeutic drugs are used in combination to target multiple mechanisms involved in cancer cell survival and proliferation. Carriers are developed to deliver drug combinations to common target tissues in optimal ratios and desirable sequences. Nanoparticles (NP) have been a popular choice for this purpose due to their ability to increase the circulation half-life and tumor accumulation of a drug. Areas covered: We review organic NP carriers based on polymers, proteins, peptides, and lipids for simultaneous delivery of multiple anticancer drugs, drug/sensitizer combinations, drug/photodynamic therapy or drug/photothermal therapy combinations, and drug/gene therapeutics with examples in the past three years. Sequential delivery of drug combinations, based on either sequential administration or built-in release control, is introduced with an emphasis on the mechanistic understanding of such control. Expert opinion: Recent studies demonstrate how a drug carrier can contribute to co-localizing drug combinations in optimal ratios and dosing sequences to maximize the synergistic effects. We identify several areas for improvement in future research, including the choice of drug combinations, circulation stability of carriers, spatiotemporal control of drug release, and the evaluation and clinical translation of combination delivery.
Collapse
Affiliation(s)
- Fanfei Meng
- a Department of Industrial and Physical Pharmacy , Purdue University , West Lafayette , IN , USA.,b Department of Pharmaceutics, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , China
| | - Ning Han
- a Department of Industrial and Physical Pharmacy , Purdue University , West Lafayette , IN , USA.,c Department of Pharmaceutics, School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , China
| | - Yoon Yeo
- a Department of Industrial and Physical Pharmacy , Purdue University , West Lafayette , IN , USA.,d Weldon School of Biomedical Engineering , Purdue University , West Lafayette , IN , USA
| |
Collapse
|
140
|
Xu HL, Mao KL, Huang YP, Yang JJ, Xu J, Chen PP, Fan ZL, Zou S, Gao ZZ, Yin JY, Xiao J, Lu CT, Zhang BL, Zhao YZ. Glioma-targeted superparamagnetic iron oxide nanoparticles as drug-carrying vehicles for theranostic effects. NANOSCALE 2016; 8:14222-14236. [PMID: 27396404 DOI: 10.1039/c6nr02448c] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Multifunctional nanoparticles capable of the specific delivery of therapeutics to diseased cells and the real-time imaging of these sites have the potential to improve cancer treatment through personalized therapy. In this study, we have proposed a multifunctional nanoparticle that integrate magnetic targeting, drug-carrier functionality and real-time MRI imaging capabilities in one platform for the theranostic treatment of tumors. The multifunctional nanoparticle was designed with a superparamagnetic iron oxide core and a multifunctional shell composed of PEG/PEI/polysorbate 80 (Ps 80) and was used to encapsulate DOX. DOX-loaded multifunctional nanoparticles (DOX@Ps 80-SPIONs) with a Dh of 58.0 nm, a zeta potential of 28.0 mV, and a drug loading content of 29.3% presented superior superparamagnetic properties with a saturation magnetization (Ms) of 24.1 emu g(-1). The cellular uptake of DOX@Ps 80-SPIONs by C6 cells under a magnetic field was significantly enhanced over that of free DOX in solution, resulting in stronger in vitro cytotoxicity. The real-time therapeutic outcome of DOX@Ps 80-SPIONs was easily monitored by MRI. Furthermore, the negative contrast enhancement effect of the nanoparticles was confirmed in glioma-bearing rats. Prussian blue staining and ex vivo DOX fluorescence assays showed that the magnetic Ps 80-SPIONs and encapsulated DOX were delivered to gliomas by imposing external magnetic fields, indicating effective magnetic targeting. Due to magnetic targeting and Ps 80-mediated endocytosis, DOX@Ps 80-SPIONs in the presence of a magnetic field led to the complete suppression of glioma growth in vivo at 28 days after treatment. The therapeutic mechanism of DOX@Ps 80-SPIONs acted by inducing apoptosis through the caspase-3 pathway. Finally, DOX@Ps 80-SPIONs' safety at therapeutic dosage was verified using pathological HE assays of the heart, liver, spleen, lung and kidney. Multifunctional SPIONs could be used as potential carriers for the theranostic treatment of CNS diseases.
Collapse
Affiliation(s)
- He-Lin Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Mujokoro B, Adabi M, Sadroddiny E, Adabi M, Khosravani M. Nano-structures mediated co-delivery of therapeutic agents for glioblastoma treatment: A review. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 69:1092-102. [PMID: 27612807 DOI: 10.1016/j.msec.2016.07.080] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/10/2016] [Accepted: 07/31/2016] [Indexed: 11/18/2022]
Abstract
Glioblastoma is a malignant brain tumor and leads to death in most patients. Chemotherapy is a common method for brain cancer in clinics. However, the recent advancements in the chemotherapy of brain tumors have not been efficient enough. With the advancement of nanotechnology, the used drugs can enhance chemotherapy efficiency and increase the access to brain cancers. Combination of therapeutic agents has been recently attracted great attention for glioblastoma chemotherapy. One of the early benefits of combination therapies is the high potential to provide synergistic effects and decrease adverse side effects associated with high doses of single anticancer drugs. Therefore, brain tumor treatments with combination drugs can be considered as a crucial approach for avoiding tumor growth. This review investigates current progress in nano-mediated co-delivery of therapeutic agents with focus on glioblastoma chemotherapy prognosis.
Collapse
Affiliation(s)
- Basil Mujokoro
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Adabi
- Young Researchers and Elite Club, Roudehen Branch, Islamic Azad University, Roudehen, Iran
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
142
|
Advances in Targeted Drug Delivery Approaches for the Central Nervous System Tumors: The Inspiration of Nanobiotechnology. J Neuroimmune Pharmacol 2016; 12:84-98. [DOI: 10.1007/s11481-016-9698-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 07/06/2016] [Indexed: 12/21/2022]
|
143
|
Xiao B, Ma L, Merlin D. Nanoparticle-mediated co-delivery of chemotherapeutic agent and siRNA for combination cancer therapy. Expert Opin Drug Deliv 2016; 14:65-73. [PMID: 27337289 DOI: 10.1080/17425247.2016.1205583] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Cancer is the leading cause of death worldwide. Current cancer treatments in the clinic mainly include chemotherapy, radiotherapy and surgery, with chemotherapy being the most common. Areas covered: Cancer treatments based on the single 'magic-bullet' concept are often associated with limited therapeutic efficacy, unwanted adverse effects, and drug resistance. The combination of multiple drugs is a promising strategy for effective cancer treatment due to the synergistic or additive effects. Small interfering RNA (siRNA) has the ability to knock down the expression of carcinogenic genes or drug efflux transporter genes, paving the way for cancer treatment. Treatment with both a chemotherapeutic agent and siRNA based on nanoparticle (NP)-mediated co-delivery is a promising approach for combination cancer therapy. Expert opinion: The combination of chemotherapeutic agents and siRNAs for cancer treatment offers the potential to enhance therapeutic efficacy, decrease side effects, and overcome drug resistance. Co-delivery of chemical drug and siRNA in the same NP would be much more effective in cancer therapy than application of chemical agent or siRNA alone. With the development of material science, NPs have come to be the most widely used platform for co-delivery of chemotherapeutic drugs and siRNAs.
Collapse
Affiliation(s)
- Bo Xiao
- a Institute for Clean Energy and Advanced Materials , Faculty for Materials and Energy, Southwest University , Chongqing , P. R. China.,b Center for Diagnostics and Therapeutics, Institute for Biomedical Sciences , Georgia State University , Atlanta , GA , USA
| | - Lijun Ma
- a Institute for Clean Energy and Advanced Materials , Faculty for Materials and Energy, Southwest University , Chongqing , P. R. China
| | - Didier Merlin
- b Center for Diagnostics and Therapeutics, Institute for Biomedical Sciences , Georgia State University , Atlanta , GA , USA.,c Veterans Affairs Medical Center , Decatur , GA , USA
| |
Collapse
|
144
|
Gao H. Progress and perspectives on targeting nanoparticles for brain drug delivery. Acta Pharm Sin B 2016; 6:268-86. [PMID: 27471668 PMCID: PMC4951594 DOI: 10.1016/j.apsb.2016.05.013] [Citation(s) in RCA: 294] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 02/06/2023] Open
Abstract
Due to the ability of the blood-brain barrier (BBB) to prevent the entry of drugs into the brain, it is a challenge to treat central nervous system disorders pharmacologically. The development of nanotechnology provides potential to overcome this problem. In this review, the barriers to brain-targeted drug delivery are reviewed, including the BBB, blood-brain tumor barrier (BBTB), and nose-to-brain barrier. Delivery strategies are focused on overcoming the BBB, directly targeting diseased cells in the brain, and dual-targeted delivery. The major concerns and perspectives on constructing brain-targeted delivery systems are discussed.
Collapse
|
145
|
Malhotra M, Toulouse A, Godinho BMDC, Mc Carthy DJ, Cryan JF, O'Driscoll CM. RNAi therapeutics for brain cancer: current advancements in RNAi delivery strategies. MOLECULAR BIOSYSTEMS 2016; 11:2635-57. [PMID: 26135606 DOI: 10.1039/c5mb00278h] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Malignant primary brain tumors are aggressive cancerous cells that invade the surrounding tissues of the central nervous system. The current treatment options for malignant brain tumors are limited due to the inability to cross the blood-brain barrier. The advancements in current research has identified and characterized certain molecular markers that are essential for tumor survival, progression, metastasis and angiogenesis. These molecular markers have served as therapeutic targets for the RNAi based therapies, which enable site-specific silencing of the gene responsible for tumor proliferation. However, to bring about therapeutic success, an efficient delivery carrier that can cross the blood-brain barrier and reach the targeted site is essential. The current review focuses on the potential of targeted, non-viral and viral particles containing RNAi therapeutic molecules as delivery strategies specifically for brain tumors.
Collapse
Affiliation(s)
- Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | | | | | | | | | | |
Collapse
|
146
|
Li T, Shen X, Geng Y, Chen Z, Li L, Li S, Yang H, Wu C, Zeng H, Liu Y. Folate-Functionalized Magnetic-Mesoporous Silica Nanoparticles for Drug/Gene Codelivery To Potentiate the Antitumor Efficacy. ACS APPLIED MATERIALS & INTERFACES 2016; 8:13748-58. [PMID: 27191965 DOI: 10.1021/acsami.6b02963] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
An appropriate codelivery system for chemotherapeutic agents and nucleic acid drugs will provide a more efficacious approach for the treatment of cancer. Combining gene therapy with chemotherapeutics in a single delivery system is more effective than individual delivery systems carrying either gene or drug. In this work, we developed folate (FA) receptor targeted magnetic-mesoporous silica nanoparticles for the codelivery of VEGF shRNA and doxorubicin (DOX) (denoted as M-MSN(DOX)/PEI-FA/VEGF shRNA). Our data showed that M-MSN(DOX)/PEI-FA could strongly condense VEGF shRNA at weight ratios of 30:1, and possesses higher stability against DNase I digestion and sodium heparin. In vitro antitumor activity assays revealed that HeLa cell growth was significantly inhibited. The intracellular accumulation of DOX by confocal microscopy and fluorescence spectrophotometry showed that M-MSN(DOX)/PEI-FA were more easily taken up than nontargeted M-MSN(DOX). Quantitative PCR and ELISA data revealed that M-MSN/PEI-FA/VEGF shRNA induced a significant decrease in VEGF expression as compared to cells treated with either the control or other complexes. The invasion and migration phenotypes of the HUVECs were significantly decrease after coculture with MSN/PEI-FA/VEGF shRNA nanocomplexes-treated HeLa cells. The approach provides a potential strategy to treat cancer by a singular nanoparticle delivery system.
Collapse
Affiliation(s)
- Tingting Li
- Department of Biophysics, School of Life Science and Technology, ‡Center for Information in Medicine, University of Electronic Science and Technology of China , Chengdu 610054, Sichuan, People's Republic of China
| | - Xue Shen
- Department of Biophysics, School of Life Science and Technology, ‡Center for Information in Medicine, University of Electronic Science and Technology of China , Chengdu 610054, Sichuan, People's Republic of China
| | - Yue Geng
- Department of Biophysics, School of Life Science and Technology, ‡Center for Information in Medicine, University of Electronic Science and Technology of China , Chengdu 610054, Sichuan, People's Republic of China
| | - Zhongyuan Chen
- Department of Biophysics, School of Life Science and Technology, ‡Center for Information in Medicine, University of Electronic Science and Technology of China , Chengdu 610054, Sichuan, People's Republic of China
| | - Li Li
- Department of Biophysics, School of Life Science and Technology, ‡Center for Information in Medicine, University of Electronic Science and Technology of China , Chengdu 610054, Sichuan, People's Republic of China
| | - Shun Li
- Department of Biophysics, School of Life Science and Technology, ‡Center for Information in Medicine, University of Electronic Science and Technology of China , Chengdu 610054, Sichuan, People's Republic of China
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology, ‡Center for Information in Medicine, University of Electronic Science and Technology of China , Chengdu 610054, Sichuan, People's Republic of China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, ‡Center for Information in Medicine, University of Electronic Science and Technology of China , Chengdu 610054, Sichuan, People's Republic of China
| | - Hongjuan Zeng
- Department of Biophysics, School of Life Science and Technology, ‡Center for Information in Medicine, University of Electronic Science and Technology of China , Chengdu 610054, Sichuan, People's Republic of China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, ‡Center for Information in Medicine, University of Electronic Science and Technology of China , Chengdu 610054, Sichuan, People's Republic of China
| |
Collapse
|
147
|
Chen D, Li B, Cai S, Wang P, Peng S, Sheng Y, He Y, Gu Y, Chen H. Dual targeting luminescent gold nanoclusters for tumor imaging and deep tissue therapy. Biomaterials 2016; 100:1-16. [PMID: 27236844 DOI: 10.1016/j.biomaterials.2016.05.017] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 05/12/2016] [Accepted: 05/14/2016] [Indexed: 11/26/2022]
Abstract
Dual targeting towards both extracellular and intracellular receptors specific to tumor is a significant approach for cancer diagnosis and therapy. In the present study, a novel nano-platform (AuNC-cRGD-Apt) with dual targeting function was initially established by conjugating gold nanocluster (AuNC) with cyclic RGD (cRGD) that is specific to αvβ3integrins over-expressed on the surface of tumor tissues and aptamer AS1411 (Apt) that is of high affinity to nucleolin over-expressed in the cytoplasm and nucleus of tumor cells. Then, AuNC-cRGD-Apt was further functionalized with near infrared (NIR) fluorescence dye (MPA), giving a NIR fluorescent dual-targeting probe AuNC-MPA-cRGD-Apt. AuNC-MPA-cRGD-Apt displays low cytotoxicity and favorable tumor-targeting capability at both in vitro and in vivo level, suggesting its clinical potential for tumor imaging. Additionally, Doxorubicin (DOX), a widely used clinical chemotherapeutic drug that kill cancer cells by intercalating DNA in cellular nucleus, was immobilized onto AuNC-cRGD-Apt forming a pro-drug, AuNC-DOX-cRGD-Apt. The enhanced tumor affinity, deep tumor penetration and improved anti-tumor activity of this pro-drug were demonstrated in different tumor cell lines, tumor spheroid and tumor-bearing mouse models. Results in this study suggest not only the prospect of non-toxic AuNC modified with two targeting ligands for tumor targeted imaging, but also confirm the promising future of dual targeting AuNC as a core for the design of prodrug in the field of cancer therapy.
Collapse
Affiliation(s)
- Dan Chen
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Bowen Li
- Department of Bioengineering, University of Washington, Seattle, USA
| | - Songhua Cai
- Nanjing University Sub-Atomic Resolution Electron Microscopy Laboratory, College of Engineering and Applied Sciences, Nanjing University, China
| | - Peng Wang
- Nanjing University Sub-Atomic Resolution Electron Microscopy Laboratory, College of Engineering and Applied Sciences, Nanjing University, China
| | - Shuwen Peng
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Yuanzhi Sheng
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Yuanyuan He
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Yueqing Gu
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China.
| | - Haiyan Chen
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China.
| |
Collapse
|
148
|
Hou J, Diao Y, Li W, Yang Z, Zhang L, Chen Z, Wu Y. RGD peptide conjugation results in enhanced antitumor activity of PD0325901 against glioblastoma by both tumor-targeting delivery and combination therapy. Int J Pharm 2016; 505:329-40. [PMID: 27085642 DOI: 10.1016/j.ijpharm.2016.04.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/28/2016] [Accepted: 04/11/2016] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is the most aggressive tumor type in the central nervous system. Both tumor-targeting drug delivery and combination therapy of multiple therapeutic agents with distinct mechanisms are important for GBM treatment. We combined these two strategies and developed a new platform of peptide-drug conjugate (RGD-PEG-Suc-PD0325901, W22) for tumor-targeting delivery using a combination of PD0325901 (a MEK1/2 inhibitor) and RGD peptide. In the present study, the combination of PD0325901 and RGD peptide strongly inhibited U87MG model in vitro and in vivo. This inhibition contributed to synergistic suppression of cell proliferation by blocking ERK pathway activity and cell migration. Modified by conjugation strategy, their conjugate W22 enhanced PD0325901 delivery to GBM cells by receptor mediated cellular internalization. W22 showed great superiority in targeting to U87MG xenografted tumors and strong anti-tumor efficacy based on ERK pathway inhibition and tumor-targeted delivery in vitro and in vivo. Moreover, W22 was stable in serum and able to release PD0325901 in the enzymatic environment. These data indicated that the RGD-PEG-Suc-PD0325901 conjugate provided a strategy for effective delivery of PD0325901 and RGD peptide into the GBM cells and inhibition of tumor growth in a synergistic manner.
Collapse
Affiliation(s)
- Jianjun Hou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Yiping Diao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Wei Li
- Department of Chemistry, Renmin University of China, Beijing 100872, PR China
| | - Zhenjun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Zili Chen
- Department of Chemistry, Renmin University of China, Beijing 100872, PR China.
| | - Yun Wu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
149
|
Wang SH, Lee ACL, Chen IJ, Chang NC, Wu HC, Yu HM, Chang YJ, Lee TW, Yu JC, Yu AL, Yu J. Structure-based optimization of GRP78-binding peptides that enhances efficacy in cancer imaging and therapy. Biomaterials 2016; 94:31-44. [PMID: 27088408 DOI: 10.1016/j.biomaterials.2016.03.050] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 01/29/2023]
Abstract
It is more challenging to design peptide drugs than small molecules through molecular docking and in silico analysis. Here, we developed a structure-based approach with various computational and analytical techniques to optimize cancer-targeting peptides for molecular imaging and therapy. We first utilized a peptide-binding protein database to identify GRP78, a specific cancer cell-surface marker, as a target protein for the lead, L-peptide. Subsequently, we used homologous modeling and molecular docking to identify a peptide-binding domain within GRP78 and optimized a series of peptides with a new protein-ligand scoring program, HotLig. Binding of these peptides to GRP78 was confirmed using an oriented immobilization technique for the Biacore system. We further examined the ability of the peptides to target cancer cells through in vitro binding studies with cell lines and clinical cancer specimens, and in vivo tumor imaging and targeted chemotherapeutic studies. MicroSPECT/CT imaging revealed significantly greater uptake of (188)Re-liposomes linked to these peptides as compared with non-targeting (188)Re-liposomes. Conjugation with these peptides also significantly increased the therapeutic efficacy of Lipo-Dox. Notably, peptide-conjugated Lipo-Dox significantly reduced stem-cell subpopulation in xenografts of breast cancer. The structure-based optimization strategy for peptides described here may be useful for developing peptide drugs for cancer imaging and therapy.
Collapse
Affiliation(s)
- Sheng-Hung Wang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan 333, Taiwan
| | - Andy Chi-Lung Lee
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan 333, Taiwan
| | - I-Ju Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Nai-Chuan Chang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan 333, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan; Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hui-Ming Yu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ya-Jen Chang
- Isotope Application Division, Institute of Nuclear Energy Research, Taiwan
| | - Te-Wei Lee
- Isotope Application Division, Institute of Nuclear Energy Research, Taiwan
| | - Jyh-Cherng Yu
- General Surgery, Department of Surgery, Tri-Service General Hospital, Taipei, Taiwan
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan 333, Taiwan; Genomics Research Center, Academia Sinica, Taipei, Taiwan; Department of Pediatrics, University of California in San Diego, USA.
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan 333, Taiwan; Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
150
|
Shigehiro T, Zhai W, Vaidyanath A, Masuda J, Mizutani A, Kasai T, Murakami H, Hamada H, Salomon DS, Mikuni K, Seno Y, Mandai T, Seno M. Evaluation of glycosylated docetaxel-encapsulated liposomes prepared by remote loading under solubility gradient. J Microencapsul 2016; 33:172-82. [PMID: 26885749 DOI: 10.3109/02652048.2016.1144815] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Docetaxel comprises one of the most effective anti-cancer drugs despite of serious side effects. Liposomes encapsulation is practically feasible to deliver the drug. However, due to the significant hydrophobicity, docetaxel will be integrated into the lipid bilayer resulting in poor encapsulation capacity. Here, we evaluated a remote loading strategy using a solubility gradient made between the two solvents for 7-glucosyloxyacetyldocetaxel, which has enhanced water solubility of docetaxel with a coupled glucose moiety. Therefore, 7-glucosyloxyacetyldocetaxel was more effectively encapsulated into liposomes with 71.0% of encapsulation efficiency than docetaxel. While 7-glucosyloxyacetyldocetaxel exhibited 90.9% of tubulin stabilisation activity of docetaxel, 7-glucosyloxyacetyldocetaxel encapsulated in liposomes significantly inhibited the growth of tumour in vivo with side effects less than unencapsulated drug. Collectively, the encapsulation of 7-glucosyloxyacetyldocetaxel into liposomes by remote loading under the solubility gradient is considered to be a promising application to prepare practical drug delivery system.
Collapse
Affiliation(s)
- Tsukasa Shigehiro
- a Department of Medical Bioengineering, Graduate School of Natural Science and Technology , Okayama University , Okayama , Japan ;,b Japan Society for the Promotion of Science , Tokyo , Japan
| | - Wenjia Zhai
- a Department of Medical Bioengineering, Graduate School of Natural Science and Technology , Okayama University , Okayama , Japan
| | - Arun Vaidyanath
- a Department of Medical Bioengineering, Graduate School of Natural Science and Technology , Okayama University , Okayama , Japan
| | - Junko Masuda
- a Department of Medical Bioengineering, Graduate School of Natural Science and Technology , Okayama University , Okayama , Japan
| | - Akifumi Mizutani
- a Department of Medical Bioengineering, Graduate School of Natural Science and Technology , Okayama University , Okayama , Japan
| | - Tomonari Kasai
- a Department of Medical Bioengineering, Graduate School of Natural Science and Technology , Okayama University , Okayama , Japan
| | - Hiroshi Murakami
- a Department of Medical Bioengineering, Graduate School of Natural Science and Technology , Okayama University , Okayama , Japan
| | - Hiroki Hamada
- c Faculty of Science , Okayama University of Science , Okayama , Japan
| | - David S Salomon
- d Mouse Cancer Genetics Program , National Cancer Institute , Frederick , MD , USA
| | | | - Yuhki Seno
- f Faculty of Life Science , Kurashiki University of Science and the Arts , Kurashiki , Japan
| | - Tadakatsu Mandai
- f Faculty of Life Science , Kurashiki University of Science and the Arts , Kurashiki , Japan
| | - Masaharu Seno
- a Department of Medical Bioengineering, Graduate School of Natural Science and Technology , Okayama University , Okayama , Japan
| |
Collapse
|