51
|
Translating the role of osteogenic-angiogenic coupling in bone formation: Highly efficient chitosan-pDNA activated scaffolds can accelerate bone regeneration in critical-sized bone defects. Biomaterials 2017; 149:116-127. [DOI: 10.1016/j.biomaterials.2017.09.036] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/12/2017] [Accepted: 09/27/2017] [Indexed: 12/31/2022]
|
52
|
Li K, Nejadnik H, Daldrup-Link HE. Next-generation superparamagnetic iron oxide nanoparticles for cancer theranostics. Drug Discov Today 2017; 22:1421-1429. [PMID: 28454771 PMCID: PMC5610947 DOI: 10.1016/j.drudis.2017.04.008] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/03/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022]
Abstract
Superparamagnetic iron oxide (SPIO) nanoparticles have been intensively studied for the development of contrast agents in MRI. First-generation SPIO nanoparticles had diagnostic capabilities only, whereas a new generation of SPIO nanoparticle has multifunctional characteristics for combined therapeutic and diagnostic applications. These theranostic nanoparticles hold great potential for image-guided cancer therapies. In particular, polymer-coated theranostic SPIO nanoparticles have enjoyed increasing attention as a result of good biocompatibility, biodegradability and versatile functionality endowed by polymeric matrices. This review provides an overview of recently developed polymer-coated multifunctional SPIO nanoparticles for cancer theranostics and discusses current challenges and future perspectives.
Collapse
Affiliation(s)
- Kai Li
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA, USA.
| | - Hossein Nejadnik
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA, USA
| | - Heike E Daldrup-Link
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
53
|
Ding X, Su Y, Wang C, Zhang F, Chen K, Wang Y, Li M, Wang W. Synergistic Suppression of Tumor Angiogenesis by the Co-delivering of Vascular Endothelial Growth Factor Targeted siRNA and Candesartan Mediated by Functionalized Carbon Nanovectors. ACS APPLIED MATERIALS & INTERFACES 2017; 9:23353-23369. [PMID: 28617574 DOI: 10.1021/acsami.7b04971] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Single-walled carbon nanotubes (SWNTs) with unique physicochemical properties have exhibited promising biomedical applications as drug and gene carriers. In this study, polyethylenimine (PEI)-modified SWNT conjugates linked with candesartan (CD) were developed to deliver vascular endothelial growth factor (VEGF)-targeted siRNA (siVEGF) for the synergistic and targeted treatment of tumor angiogenesis. The characterization results revealed that SWNT-PEI-CD conjugates were successfully synthesized and exhibited desirable dispersibility and superior stability. Confocal laser scanning microscopy (CLSM) and flow cytometry (FCM) results showed that SWNT-PEI-CD/siVEGF complexes could achieve high cellular uptake and specific intracellular distribution of siRNA in AT1R overexpressed PANC-1 cells. Strong down-regulation of VEGF was also verified by qualitative real-time polymerase chain reaction, enzyme-linked immunosorbent assay, and Western blot in complex-treated PANC-1 cells. The in vitro angiogenesis assay showed that SWNT-PEI-CD/siVEGF complexes highly inhibited tube formation of human umbilical vein endothelial cells. Furthermore, in vivo observation in PANC-1 xenografted nude mice demonstrated that SWNT-PEI-CD/siVEGF complexes exhibited significant distribution at tumor sites and caused obvious inhibition of tumor growth and tumor-associated angiogenesis repression induced by the drug combination of CD and siVEGF. Finally, a WST-1 assay indicated that SWNT-PEI-CD possessed low cytotoxicity, and a hemolysis test showed good biocompatibility of SWNT-PEI-CD. Hematological and histological analyses confirmed that SWNT-PEI-CD/siVEGF complexes did not cause any obvious toxic effects to blood and major organs. These findings suggested that the SWNT-PEI-CD/siVEGF co-delivery system with tumor-targeting specificity, improved endosomal escaping properties, and collaboration of angiogenesis inhibition could be a prospective method for efficient tumor antiangiogenic therapy.
Collapse
Affiliation(s)
- Xuefang Ding
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
- School of Pharmaceutical Science, Nanjing Tech University , 30 South Puzhu Road, Nanjing 211816, China
| | - Yujie Su
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Cheng Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Fangrong Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Kerong Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Yu Wang
- Department of Pharmacology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University , 140 Hanzhong Road, Nanjing 210029, China
| | - Min Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| |
Collapse
|
54
|
Jiang Y, Sprouse D, Laaser JE, Dhande Y, Reineke TM, Lodge TP. Complexation of Linear DNA and Poly(styrenesulfonate) with Cationic Copolymer Micelles: Effect of Polyanion Flexibility. J Phys Chem B 2017; 121:6708-6720. [DOI: 10.1021/acs.jpcb.7b03732] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Yaming Jiang
- Department of Chemical Engineering & Materials Science, University of Minnesota, 421 Washington Ave. SE, Minneapolis, Minnesota 55455, United States
| | - Dustin Sprouse
- Department
of Chemistry, University of Minnesota, 207 Pleasant St. SE, Minneapolis, Minnesota 55455, United States
| | - Jennifer E. Laaser
- Department
of Chemistry, University of Minnesota, 207 Pleasant St. SE, Minneapolis, Minnesota 55455, United States
| | - Yogesh Dhande
- Department of Chemical Engineering & Materials Science, University of Minnesota, 421 Washington Ave. SE, Minneapolis, Minnesota 55455, United States
| | - Theresa M. Reineke
- Department
of Chemistry, University of Minnesota, 207 Pleasant St. SE, Minneapolis, Minnesota 55455, United States
| | - Timothy P. Lodge
- Department of Chemical Engineering & Materials Science, University of Minnesota, 421 Washington Ave. SE, Minneapolis, Minnesota 55455, United States
- Department
of Chemistry, University of Minnesota, 207 Pleasant St. SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
55
|
Huang X, Lee RJ, Qi Y, Li Y, Lu J, Meng Q, Teng L, Xie J. Microfluidic hydrodynamic focusing synthesis of polymer-lipid nanoparticles for siRNA delivery. Oncotarget 2017; 8:96826-96836. [PMID: 29228574 PMCID: PMC5722526 DOI: 10.18632/oncotarget.18281] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/01/2017] [Indexed: 12/12/2022] Open
Abstract
Small interfering RNAs (siRNAs) are promising as therapeutics for intractable diseases such as cancer. However, efficient and safe delivery of siRNAs in vivo remains a challenge. Polymer-lipid hybrid nanoparticles (P/LNPs) have been evaluated for therapeutic delivery of siRNA. In this study, a microfluidic hydrodynamic focusing (MF) system was used to prepare P/LNPs loaded with VEGF siRNA. P/LNPs made by MF were smaller in particle size and had narrower size distribution compared to P/LNPs formed by bulk mixing (BM). MF-synthesized P/LNPs demonstrated low vehicle cytotoxicity and potent tumor cell inhibition in vitro. In addition, P/LNPs produced by the microfluidic chip exhibited prolonged blood circulation and increased AUC after i.v. injection compared to free siRNA. Furthermore, P/LNPs synthesized by MF induced greater down-regulation of VEGF mRNA and protein levels as well as greater tumor inhibition in a xenograft tumor model. Taken together, P/LNPs prepared by MF have been shown to be an effective and safe therapeutic siRNA delivery system for cancer treatment both in vitro and in vivo.
Collapse
Affiliation(s)
- Xueqin Huang
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China.,Department of Chemistry and Pharmacy, Zhuhai College of Jilin University, Zhuhai, Guangdong, 519041, China
| | - Robert J Lee
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China.,Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA
| | - Yuhang Qi
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| | - Yujing Li
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| | - Jiahui Lu
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| | - Qingfan Meng
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| | - Jing Xie
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| |
Collapse
|
56
|
Wang G, Gao X, Gu G, Shao Z, Li M, Wang P, Yang J, Cai X, Li Y. Polyethylene glycol-poly(ε-benzyloxycarbonyl-l-lysine)-conjugated VEGF siRNA for antiangiogenic gene therapy in hepatocellular carcinoma. Int J Nanomedicine 2017; 12:3591-3603. [PMID: 28533682 PMCID: PMC5431695 DOI: 10.2147/ijn.s131078] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
A polyethylene glycol-poly(ε-benzyloxycarbonyl-l-lysine) (PEG-SS-PLL) block copolymer based on a disulfide-linked, novel biodegradable catiomer bearing a PEG-sheddable shell was developed to avoid "PEG dilemma" in nanoparticle intracellular tracking of PEG-PLL where PEG was nondegradable. However, PEG-SS-PLL catiomers have not been used to deliver small interfering VEGF RNA (siVEGF) in antiangiogenesis gene therapy. In this study, we aimed to investigate whether this novel biodegradable catiomer can deliver siVEGF into cancer cells and at the same time have an antitumor effect in a xenograft mouse model. It was found that PEG-SS-PLL efficiently delivered siVEGF with negligible cytotoxicity, and significantly decreased the expression of VEGF at both the messenger-RNA and protein levels both in vitro and in vivo, and thus tumor growth was inhibited. Our findings demonstrated that PEG-SS-PLL/siVEGF could potentially be applied to antiangiogenesis gene therapy for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Gangmin Wang
- Department of Urology, Huashan Hospital, Fudan University
| | - XiaoLong Gao
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai
| | - GuoJun Gu
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai
| | - ZhiHong Shao
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai
| | - MingHua Li
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai
| | - PeiJun Wang
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai
| | - JianRong Yang
- Department of Hepatobiliary Surgery, Third People's Hospital of Guangxi Zhuang Autonomous Region, Nanning
| | - XiaoJun Cai
- Institute for Advanced Materials and Nano Biomedicine, School of Material Science and Engineering, Tongji University, Shanghai, People's Republic of China
| | - YongYong Li
- Institute for Advanced Materials and Nano Biomedicine, School of Material Science and Engineering, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
57
|
Lee JH, Ku SH, Kim MJ, Lee SJ, Kim HC, Kim K, Kim SH, Kwon IC. Rolling circle transcription-based polymeric siRNA nanoparticles for tumor-targeted delivery. J Control Release 2017; 263:29-38. [PMID: 28373128 DOI: 10.1016/j.jconrel.2017.03.390] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/09/2017] [Accepted: 03/26/2017] [Indexed: 02/06/2023]
Abstract
RNA, one of the major biological macromolecules, has been considered as an attractive building material for bottom-up fabrication of nanostructures in the past few decades due to advancements in RNA biology, RNA chemistry and RNA nanotechnology. Most recently, an isothermal enzymatic nucleic acid amplification method termed rolling circle transcription (RCT), which achieves a large-scale synthesis of RNA nanostructures, has emerged as one of fascinating techniques for RNAi-based therapies. Herein, we proposed a newly designed RCT method for synthesis of polymeric siRNA nanoflower, referred to 'RCT and annealing-generated polymeric siRNA (RAPSI)': (1) Amplification of the antisense strand of siRNA via RCT process and (2) annealing of chimeric sense strand containing 3'-terminal DNA nucleotides that provide enzyme cleavage sites. To verify its potentials in RNAi-based cancer therapy, the newly designed RAPSI nanoflower was further complexed with glycol chitosan (GC) derivatives, and systemically delivered to PC-3 xenograft tumors. The resultant RAPSI nanoparticles exhibited the improved particle stability against polyanion competition or nuclease attack. When the RAPSI nanoparticles reached to the cytoplasmic region, active mono siRNA was liberated and significantly down-regulated the expression of target VEGF gene in PC-3 cells. Excellent tumor-homing efficacy and anti-tumor effects of the RAPSI nanoparticles were further demonstrated. Overall, the proposed RCT-based polymeric siRNA nanoflower formulation can provide a new platform technology that allows further functional modifications via an advanced annealing method for systemic cancer RNAi therapy.
Collapse
Affiliation(s)
- Jae Hyeop Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, Republic of Korea
| | - Sook Hee Ku
- Technology Convergence R&D Group, Korea Institute of Industrial Technology, Daegu, Republic of Korea
| | - Min Ju Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - So Jin Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hyun Cheol Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, Republic of Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
58
|
Jedrzejczyk D, Gendaszewska-Darmach E, Pawlowska R, Chworos A. Designing synthetic RNA for delivery by nanoparticles. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2017; 29:123001. [PMID: 28004640 DOI: 10.1088/1361-648x/aa5561] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The rapid development of synthetic biology and nanobiotechnology has led to the construction of various synthetic RNA nanoparticles of different functionalities and potential applications. As they occur naturally, nucleic acids are an attractive construction material for biocompatible nanoscaffold and nanomachine design. In this review, we provide an overview of the types of RNA and nucleic acid's nanoparticle design, with the focus on relevant nanostructures utilized for gene-expression regulation in cellular models. Structural analysis and modeling is addressed along with the tools available for RNA structural prediction. The functionalization of RNA-based nanoparticles leading to prospective applications of such constructs in potential therapies is shown. The route from the nanoparticle design and modeling through synthesis and functionalization to cellular application is also described. For a better understanding of the fate of targeted RNA after delivery, an overview of RNA processing inside the cell is also provided.
Collapse
Affiliation(s)
- Dominika Jedrzejczyk
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland
| | | | | | | |
Collapse
|
59
|
SiRNA-mediated in vivo gene knockdown by acid-degradable cationic nanohydrogel particles. J Control Release 2017; 248:10-23. [DOI: 10.1016/j.jconrel.2016.12.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/23/2016] [Accepted: 12/06/2016] [Indexed: 01/22/2023]
|
60
|
Palanca-Wessels MC, Booth GC, Convertine AJ, Lundy BB, Berguig GY, Press MF, Stayton PS, Press OW. Antibody targeting facilitates effective intratumoral siRNA nanoparticle delivery to HER2-overexpressing cancer cells. Oncotarget 2017; 7:9561-75. [PMID: 26840082 PMCID: PMC4891060 DOI: 10.18632/oncotarget.7076] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 01/19/2016] [Indexed: 11/25/2022] Open
Abstract
The therapeutic potential of RNA interference (RNAi) has been limited by inefficient delivery of short interfering RNA (siRNA). Tumor-specific recognition can be effectively achieved by antibodies directed against highly expressed cancer cell surface receptors. We investigated the utility of linking an internalizing streptavidin-conjugated HER2 antibody to an endosome-disruptive biotinylated polymeric nanocarrier to improve the functional cytoplasmic delivery of siRNA in breast and ovarian cancer cells in vitro and in an intraperitoneal ovarian cancer xenograft model in vivo, yielding an 80% reduction of target mRNA and protein levels with sustained repression for at least 96 hours. RNAi-mediated site specific cleavage of target mRNA was demonstrated using the 5′ RLM-RACE (RNA ligase mediated-rapid amplification of cDNA ends) assay. Mice bearing intraperitoneal human ovarian tumor xenografts demonstrated increased tumor accumulation of Cy5.5 fluorescently labeled siRNA and 70% target gene suppression after treatment with HER2 antibody-directed siRNA nanocarriers. Detection of the expected mRNA cleavage product by 5′ RLM-RACE assay confirmed that suppression occurs via the expected RNAi pathway. Delivery of siRNA via antibody-directed endosomolytic nanoparticles may be a promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Maria C Palanca-Wessels
- Clinical Research Division and Center for Intracellular Delivery of Biologics, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Medicine, Hematology Division, University of Washington, Seattle, WA, USA
| | - Garrett C Booth
- Clinical Research Division and Center for Intracellular Delivery of Biologics, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Anthony J Convertine
- Department of Bioengineering and Center for Intracellular Delivery of Biologics, University of Washington, Seattle, WA, USA
| | - Brittany B Lundy
- Department of Bioengineering and Center for Intracellular Delivery of Biologics, University of Washington, Seattle, WA, USA
| | - Geoffrey Y Berguig
- Department of Bioengineering and Center for Intracellular Delivery of Biologics, University of Washington, Seattle, WA, USA
| | - Michael F Press
- Department of Pathology, University of Southern California, Los Angeles, CA, USA
| | - Patrick S Stayton
- Department of Bioengineering and Center for Intracellular Delivery of Biologics, University of Washington, Seattle, WA, USA
| | - Oliver W Press
- Clinical Research Division and Center for Intracellular Delivery of Biologics, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Bioengineering and Center for Intracellular Delivery of Biologics, University of Washington, Seattle, WA, USA
| |
Collapse
|
61
|
Recent advances in the design, development, and targeting mechanisms of polymeric micelles for delivery of siRNA in cancer therapy. Prog Polym Sci 2017. [DOI: 10.1016/j.progpolymsci.2016.09.008] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
62
|
Kim D, Ku SH, Kim H, Jeong JH, Lee M, Kwon IC, Choi D, Kim SH. Simultaneous regulation of apoptotic gene silencing and angiogenic gene expression for myocardial infarction therapy: Single-carrier delivery of SHP-1 siRNA and VEGF-expressing pDNA. J Control Release 2016; 243:182-194. [DOI: 10.1016/j.jconrel.2016.10.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/23/2016] [Accepted: 10/14/2016] [Indexed: 02/06/2023]
|
63
|
Ho W, Zhang XQ, Xu X. Biomaterials in siRNA Delivery: A Comprehensive Review. Adv Healthc Mater 2016; 5:2715-2731. [PMID: 27700013 DOI: 10.1002/adhm.201600418] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/07/2016] [Indexed: 01/31/2023]
Abstract
With the dearth of effective treatment options for prominent diseases including Ebola and cancer, RNA interference (RNAi), a sequence-specific mechanism for genetic regulation that can silence nearly any gene, holds the promise of unlimited potential in treating illness ever since its discovery in 1999. Given the large size, unstable tertiary structure in physiological conditions and negative charge of small interfering RNAs (siRNAs), the development of safe and effective delivery vehicles is of critical importance in order to drive the widespread use of RNAi therapeutics into clinical settings. Immense amounts of time and billions of dollars have been devoted into the design of novel and diverse delivery strategies, and there are a handful of delivery systems that have been successfully translated into clinic. This review provides an introduction to the in vivo barriers that need to be addressed by siRNA delivery systems. We also discuss the progress up to the most effective and clinically advanced siRNA delivery systems including liposomal, polymeric and siRNA conjugate delivery systems, as well as their design to overcome the challenges.
Collapse
Affiliation(s)
- William Ho
- Department of Chemical, Biological and Pharmaceutical Engineering; Newark School of Engineering; New Jersey Institute of Technology; Newark NJ 07102 USA
| | - Xue-Qing Zhang
- Department of Chemical, Biological and Pharmaceutical Engineering; Newark School of Engineering; New Jersey Institute of Technology; Newark NJ 07102 USA
| | - Xiaoyang Xu
- Department of Chemical, Biological and Pharmaceutical Engineering; Newark School of Engineering; New Jersey Institute of Technology; Newark NJ 07102 USA
| |
Collapse
|
64
|
Chemical and structural modifications of RNAi therapeutics. Adv Drug Deliv Rev 2016; 104:16-28. [PMID: 26549145 DOI: 10.1016/j.addr.2015.10.015] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 10/14/2015] [Accepted: 10/23/2015] [Indexed: 12/25/2022]
Abstract
Small interfering RNA (siRNA), a 21-23nt double-stranded RNA responsible for post-transcriptional gene silencing, has attracted great interests as promising genomic drugs, due to its strong ability to silence target genes in a sequence-specific manner. Despite high silencing efficiency and on-target specificity, the clinical translation of siRNA has been hindered by its inherent features: poor intracellular delivery, limited blood stability, unpredictable immune responses and unwanted off-targeting effects. To overcome these hindrances, researchers have made various advances to modify siRNA itself and to improve its delivery. In this review paper, first we briefly discuss the innate properties and delivery barriers of siRNA. Then, we describe recent progress in (1) chemically and structurally modified siRNAs to solve their intrinsic problems and (2) siRNA delivery formulations including siRNA conjugates, polymerized siRNA, and nucleic acid-based nanoparticles to improve in vivo delivery.
Collapse
|
65
|
Wang J, Mi P, Lin G, Wáng YXJ, Liu G, Chen X. Imaging-guided delivery of RNAi for anticancer treatment. Adv Drug Deliv Rev 2016; 104:44-60. [PMID: 26805788 DOI: 10.1016/j.addr.2016.01.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 11/27/2015] [Accepted: 01/13/2016] [Indexed: 12/12/2022]
Abstract
The RNA interference (RNAi) technique is a new modality for cancer therapy, and several candidates are being tested clinically. In the development of RNAi-based therapeutics, imaging methods can provide a visible and quantitative way to investigate the therapeutic effect at anatomical, cellular, and molecular level; to noninvasively trace the distribution; to and study the biological processes in preclinical and clinical stages. Their abilities are important not only for therapeutic optimization and evaluation but also for shortening of the time of drug development to market. Typically, imaging-functionalized RNAi therapeutics delivery that combines nanovehicles and imaging techniques to study and improve their biodistribution and accumulation in tumor site has been progressively integrated into anticancer drug discovery and development processes. This review presents an overview of the current status of translating the RNAi cancer therapeutics in the clinic, a brief description of the biological barriers in drug delivery, and the roles of imaging in aspects of administration route, systemic circulation, and cellular barriers for the clinical translation of RNAi cancer therapeutics, and with partial content for discussing the safety concerns. Finally, we focus on imaging-guided delivery of RNAi therapeutics in preclinical development, including the basic principles of different imaging modalities, and their advantages and limitations for biological imaging. With growing number of RNAi therapeutics entering the clinic, various imaging methods will play an important role in facilitating the translation of RNAi cancer therapeutics from bench to bedside.
Collapse
|
66
|
Lee SH, Kang YY, Jang HE, Mok H. Current preclinical small interfering RNA (siRNA)-based conjugate systems for RNA therapeutics. Adv Drug Deliv Rev 2016; 104:78-92. [PMID: 26514375 DOI: 10.1016/j.addr.2015.10.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 09/01/2015] [Accepted: 10/12/2015] [Indexed: 01/01/2023]
Abstract
Recent promising clinical results of RNA therapeutics have drawn big attention of academia and industries to RNA therapeutics and their carrier systems. To improve their feasibility in clinics, systemic evaluations of currently available carrier systems under clinical trials and preclinical studies are needed. In this review, we focus on recent noticeable preclinical studies and clinical results regarding siRNA-based conjugates for clinical translations. Advantages and drawbacks of siRNA-based conjugates are discussed, compared to particle-based delivery systems. Then, representative siRNA-based conjugates with aptamers, peptides, carbohydrates, lipids, polymers, and nanostructured materials are introduced. To improve feasibility of siRNA conjugates in preclinical studies, several considerations for the rational design of siRNA conjugates in terms of cleavability, immune responses, multivalent conjugations, and mechanism of action are also presented. Lastly, we discuss lessons from previous preclinical and clinical studies related to siRNA conjugates and perspectives of their clinical applications.
Collapse
Affiliation(s)
- Soo Hyeon Lee
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), Zurich, Switzerland
| | - Yoon Young Kang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Hyo-Eun Jang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea.
| |
Collapse
|
67
|
Jeong EH, Kim H, Jang B, Cho H, Ryu J, Kim B, Park Y, Kim J, Lee JB, Lee H. Technological development of structural DNA/RNA-based RNAi systems and their applications. Adv Drug Deliv Rev 2016; 104:29-43. [PMID: 26494399 DOI: 10.1016/j.addr.2015.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 09/01/2015] [Accepted: 10/12/2015] [Indexed: 01/04/2023]
Abstract
RNA interference (RNAi)-based gene therapy has drawn tremendous attention due to its highly specific gene regulation by selective degradation of any target mRNA. There have been multiple reports regarding the development of various cationic materials for efficient siRNA delivery, however, many studies still suffer from the conventional delivery problems such as suboptimal transfection performance, a lack of tissue specificity, and potential cytotoxicity. Despite the huge therapeutic potential of siRNAs, conventional gene carriers have failed to guarantee successful gene silencing in vivo, thus not warranting clinical trials. The relatively short double-stranded structure of siRNAs has resulted in uncompromising delivery formulations, as well as low transfection efficiency, compared with the conventional nucleic acid drugs such as plasmid DNAs. Recent developments in structural siRNA and RNAi nanotechnology have enabled more refined and reliable in vivo gene silencing with multiple advantages over naked siRNAs. This review focuses on recent progress in the development of structural DNA/RNA-based RNAi systems and their potential therapeutic applications. In addition, an extensive list of prior reports on various RNAi systems is provided and categorized by their distinctive molecular characters.
Collapse
|
68
|
Tzeng SY, Wilson DR, Hansen SK, Quiñones-Hinojosa A, Green JJ. Polymeric nanoparticle-based delivery of TRAIL DNA for cancer-specific killing. Bioeng Transl Med 2016; 1:149-159. [PMID: 28349127 PMCID: PMC5365091 DOI: 10.1002/btm2.10019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Lack of specificity in cancer therapeutics severely limits the efficacy of many existing treatment modalities. The use of Tumor Necrosis Factor-related Apoptosis-Inducing Ligand (TRAIL) is of interest to the field due to this protein's ability to cause cell death specifically in cancer cells without harming the surrounding healthy tissue. Here, we report that polymeric nanoparticles, based on synthetic poly(beta-amino ester)s (PBAEs) and containing DNA, are able to selectively transfect cancer cells in vitro over healthy cells of the same tissue type. Moreover, PBAE-based nanoparticles containing TRAIL DNA are able to transfect several human cancer cell cultures in vitro and cause cell death. While certain cell types, including human glioblastoma (GBM), showed resistance to TRAIL, we found that the expression of TRAIL-binding surface proteins was predictive of each cell type's resistance to TRAIL therapy. We demonstrate a non-viral nanomedicine approach to cancer gene therapy that can improve cancer specificity via both biomaterial selection and through the use of cancer-targeting genetic cargo.
Collapse
Affiliation(s)
- Stephany Y Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD; The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - David R Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD; The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sarah K Hansen
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD; The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD; The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Material Science and Engineering, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
69
|
Zoheir KM, Abd-Rabou AA, Harisa GI, Kumar A, Ahmad SF, Ansari MA, Abd-Allah AR. IQGAP1 gene silencing induces apoptosis and decreases the invasive capacity of human hepatocellular carcinoma cells. Tumour Biol 2016; 37:13927-13939. [PMID: 27488117 DOI: 10.1007/s13277-016-5283-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/15/2016] [Indexed: 12/24/2022] Open
Abstract
IQ motif-containing GTPase-activating proteins (IQGAPs) belong to a conserved family, and they are involved in various intracellular processes. IQGAP1 is expressed in all cells, while IQGAP2 and IQGAP3 are mainly expressed in hepatic cells. IQGAP1 has been suggested to be an oncogene, while IQGAP2 is considered a tumor-suppressor gene. However, the relationship between RAS family genes and IQGAP genes remains unclear. We recently demonstrated this interaction in a chemically induced mouse liver cancer. In this study, IQGAP1 expression was partially silenced in human hepatocellular carcinoma (HepG2) cells. We investigated the impact of IQGAP1 silencing on the interactions of IQGAP and RAS with several apoptotic proteins, including caspase-3 (CASP3), BCL2-associated X protein (BAX), and B-cell leukemia/lymphoma 2 (BCL2). Additionally, we investigated the effects of the interactions of these genes on cell viability, proliferation, apoptosis, and invasive capacity. IQGAP1 siRNA-treated HepG2 cells showed lower invasive capacity than the control cells, and this reduction was time- and vector concentration-dependent. In addition, IQGAP1 silencing resulted in significantly lower IQGAP1 level and subsequently higher IQGAP2 and IQGAP3 expression in HepG2 cells than in the control. Flow cytometry analyses indicated that the silencing of IQGAP1 can induce early and late apoptosis in HepG2 cells. Additionally, IQGAP2, IQGAP3, CASP3, and BAX were upregulated whereas IQGAP1 and BCL2 were downregulated in the siRNA-treated cells. Furthermore, we observed that the mRNA levels of HRAS, KRAS, NRAS, and MRAS decreased upon IQGAP1 silencing. These findings indicate that IQGAP1 potentially regulates the expression of IQGAP and RAS gene families and demonstrate its regulatory role in the apoptotic network. Taken together, our findings suggest that IQGAP1 silencing plays crucial roles in the apoptosis of HepG2 cells and lowers their proliferative and invasive capacities.
Collapse
Affiliation(s)
- Khairy Ma Zoheir
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia. .,Cell Biology Department, National Research Centre, Cairo, 12622, Egypt.
| | - Ahmed A Abd-Rabou
- Hormones Department, Medical Research Division, National Research Centre, Cairo, 12622, Egypt
| | - Gamaleldin I Harisa
- Department of Pharmaceutics, College of Pharmacy, King Saud University, PO Box 11451, Riyadh, Saudi Arabia
| | - Ashok Kumar
- Vitiligo Research Chair, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh Fayaz Ahmad
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mushtaq Ahmad Ansari
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Adel R Abd-Allah
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
70
|
RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients. Oncotarget 2016; 6:24560-70. [PMID: 26009994 PMCID: PMC4695206 DOI: 10.18632/oncotarget.4183] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/02/2015] [Indexed: 12/12/2022] Open
Abstract
Purpose The miniature biodegradable implant siG12D-LODER™ was inserted into a tumor and released a siRNA drug against KRAS(G12D) along four months. This novel siRNA based drug was studied, in combination with chemotherapy, as targeted therapy for Locally Advanced Pancreatic Cancer (LAPC). Methods An open-label Phase 1/2a study in the first-line setting of patients with non-operable LAPC was initiated. In this study patients were assigned to receive a single dose of siG12D-LODERs, in three escalating dose cohorts (0.025mg, 0.75mg and 3.0mg). Gemcitabine was given on a weekly basis, following the siG12D-LODERTM insertion, until disease progression. The recommended dose was further examined with modified FOLFIRINOX. The follow up period was eight weeks and survival until death. Results Fifteen patients with LAPC were enrolled. Among the 15 treated patients, the most frequent adverse events observed were grade 1or 2 in severity (89%); five patients experienced serious adverse events (SAEs). In 12 patients analyzed by CT scans, none showed tumor progression, the majority (10/12) demonstrated stable disease and two showed partial response. Decrease in tumor marker CA19-9 was observed in 70% (7/10) of patients. Median overall survival was 15.12 months; 18 month survival was 38.5%. Conclusions The combination of siG12D-LODER™ and chemotherapy is well tolerated, safe and demonstrated a potential efficacy in patients with LAPC. NCT01188785
Collapse
|
71
|
Kim HS, Son YJ, Yoo HS. Clustering siRNA conjugates for MMP-responsive therapeutics in chronic wounds of diabetic animals. NANOSCALE 2016; 8:13236-13244. [PMID: 27251781 DOI: 10.1039/c6nr01551d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The MMP-responsive breakdown of siRNA clusters was translated to site-specific gene transfection and enhanced wound healing in diabetic ulcers. MMP-2 siRNA was chemically tethered to the end of multi-armed PEG via MMP-cleavable linkers (4PEG-siRNA) and subsequently clustered into submicron particles complexed with LPEI. 4PEG-siRNA was more tightly complexed with LPEI and the associated cluster showed higher resistance against RNase attack, in comparison to naked siRNA. Because the size of the clusters increased depending on the increase in charge ratio of LPEI to siRNA, cellular uptake of the 4PEG-siRNA/LPEI cluster was significantly attenuated due to the huge size of the cluster. However, upon MMP treatment, the cluster dissociated into smaller particles and was efficiently endocytosed by cells. An in vivo fluorescence resonance energy transfer (FRET) study also revealed that the clusters were effectively dissociated in MMP-rich environments of dorsal wounds in diabetic animals. In addition, diabetic ulcers treated with the clusters showed a faster wound closure rate and the recovered tissue expressed a larger amount of cytokeratin along with a lower expression level of MMP-2 compared to the other groups.
Collapse
Affiliation(s)
- Hye Sung Kim
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon,24341, Republic of Korea.
| | | | | |
Collapse
|
72
|
Zhang CG, Yang SD, Zhu WJ, You BG, Liu Y, Yuan ZQ, Chen WL, Li JZ, Zhou XF, Liu C, Zhang XN. Distinctive polymer micelle designed for siRNA delivery and reversal of MDR1 gene-dependent multidrug resistance. J Biomed Mater Res B Appl Biomater 2016; 105:2093-2106. [PMID: 27405391 DOI: 10.1002/jbm.b.33748] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/09/2016] [Accepted: 06/27/2016] [Indexed: 11/11/2022]
Abstract
P-glycoprotein (P-gp) plays an importantrole in multidrug resistance (MDR), proved to be one of the major obstacles in cancer chemotherapy. Cationic polymers could specifically deliver siRNA to tumor cells and thus reverse MDR by the downregulation of P-gp. In this study, a triblock copolymer micelle was prepared based on the polymer of N-succinyl-chitosan-poly-l-lysine-palmitic acid (NSC-PLL-PA) to deliver siRNA-P-gp (siRNA-micelle) or doxorubicin (Dox-micelle). The resulting micelle exhibited an efficient binding ability for siRNA and high encapsulation efficiency for Dox, with an average particle size of ∼170 nm. siRNA-micelle and Dox-micellewere instable at low pH, thereby enhancing tumor accumulation and intracellular release of the encapsulated siRNA and Dox. siRNA-micelle micelles could enhance the knockdown efficacy of siRNA by improving the transfection efficiency, downregulating P-gp expression, and passing the drug efflux transporters, thereby improving the therapeutic effects of Dox-micelle. However, P-gp could transfer from HepG2/ADM to HepG2 cells independent of the expression of mdr1, and the acquired resistance could permit tumor cells to survive and develop intrinsic P-gp-mediated resistance, thereby limiting the desired efficiency of chemotherapeutics. This study demonstrated the effectiveness of siRNA-micelle for tumor-targeted delivery, MDR reversal, and provided an effective strategy for the treatment of cancers that develop MDR. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 2093-2106, 2017.
Collapse
Affiliation(s)
- Chun-Ge Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China.,The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Shu-di Yang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Wen-Jing Zhu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Ben-Gang You
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yang Liu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Zhi-Qiang Yuan
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Wei-Liang Chen
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Ji-Zhao Li
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Xiao-Feng Zhou
- College of Radiological Medicine and Protection, Soochow University, Suzhou, People's Republic of China.,Changshu Hospital of Traditional Chinese Medicine, Changshu, People's Republic of China
| | - Chun Liu
- The Hospital of Suzhou People's Hospital Affiliated to Nanjing Medical University, Suzhou, People's Republic of China
| | - Xue-Nong Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
73
|
Raftery RM, Walsh DP, Castaño IM, Heise A, Duffy GP, Cryan SA, O'Brien FJ. Delivering Nucleic-Acid Based Nanomedicines on Biomaterial Scaffolds for Orthopedic Tissue Repair: Challenges, Progress and Future Perspectives. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:5447-5469. [PMID: 26840618 DOI: 10.1002/adma.201505088] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/27/2015] [Indexed: 06/05/2023]
Abstract
As well as acting to fill defects and allow for cell infiltration and proliferation in regenerative medicine, biomaterial scaffolds can also act as carriers for therapeutics, further enhancing their efficacy. Drug and protein delivery on scaffolds have shown potential, however, supraphysiological quantities of therapeutic are often released at the defect site, causing off-target side effects and cytotoxicity. Gene therapy involves the introduction of foreign genes into a cell in order to exert an effect; either replacing a missing gene or modulating expression of a protein. State of the art gene therapy also encompasses manipulation of the transcriptome by harnessing RNA interference (RNAi) therapy. The delivery of nucleic acid nanomedicines on biomaterial scaffolds - gene-activated scaffolds -has shown potential for use in a variety of tissue engineering applications, but as of yet, have not reached clinical use. The current state of the art in terms of biomaterial scaffolds and delivery vector materials for gene therapy is reviewed, and the limitations of current procedures discussed. Future directions in the clinical translation of gene-activated scaffolds are also considered, with a particular focus on bone and cartilage tissue regeneration.
Collapse
Affiliation(s)
- Rosanne M Raftery
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
- Drug Delivery and Advanced Materials Research Team, School of Pharmacy, Royal College of Surgeons in Ireland, 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - David P Walsh
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
- Drug Delivery and Advanced Materials Research Team, School of Pharmacy, Royal College of Surgeons in Ireland, 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Irene Mencía Castaño
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Andreas Heise
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
| | - Garry P Duffy
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Sally-Ann Cryan
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Drug Delivery and Advanced Materials Research Team, School of Pharmacy, Royal College of Surgeons in Ireland, 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| |
Collapse
|
74
|
Conjugates of small targeting molecules to non-viral vectors for the mediation of siRNA. Acta Biomater 2016; 36:21-41. [PMID: 27045350 DOI: 10.1016/j.actbio.2016.03.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/29/2016] [Accepted: 03/31/2016] [Indexed: 01/08/2023]
Abstract
UNLABELLED To use siRNA (small interfering RNA) for gene therapy, a gene delivery system is often necessary to overcome several challenging requirements including rapid excretion, low stability in blood serum, non-specific accumulation in tissues, poor cellular uptake and inefficient intracellular release. Active and/or passive targeting should help the delivery system to reach the desired tissue or cell, to be internalized, and to deliver siRNA to the cytoplasm so that siRNA can inhibit protein synthesis. This review covers conjugates of small targeting molecules and non-viral delivery systems for the mediation of siRNA, with a focus on their transfection properties in order to help the development of new and efficient siRNA delivery systems, as the therapeutic solutions of tomorrow. STATEMENT OF SIGNIFICANCE The delivery of siRNA into cells or tissues remains to be a challenge for its applications, an alternative strategy for siRNA delivery systems is direct conjugation of non-viral vectors with targeting moieties for cellular delivery. In comparison to macromolecules, small targeting molecules have attracted great attention due to their many potential advantages including significant simplicity and ease of production, good repeatability and biodegradability. This review will focus on the most recent advances in the delivery of siRNA using conjugates of small targeting molecules and non-viral delivery systems. Based the editor's suggestions, we hope the revised manuscript could provide more profound understanding to the conjugates of targeting molecules to vectors for mediation of siRNA.
Collapse
|
75
|
Lee SY, Yang CY, Peng CL, Wei MF, Chen KC, Yao CJ, Shieh MJ. A theranostic micelleplex co-delivering SN-38 and VEGF siRNA for colorectal cancer therapy. Biomaterials 2016; 86:92-105. [PMID: 26896610 DOI: 10.1016/j.biomaterials.2016.01.068] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 01/31/2016] [Indexed: 02/08/2023]
Abstract
The development of an efficient colorectal cancer therapy is currently a public health priority. In the present work, we proposed a multifunctional theranostic micellar drug delivery system utilizing cationic PDMA-block-poly(ε-caprolactone) (PDMA-b-PCL) micelles as nanocarriers of SN-38 (7-ethyl-10-hydroxycamptothecin), ultra-small superparamagnetic iron oxide nanoparticles (USPIO), and small interfering RNA (siRNA) that targets human vascular endothelial growth factor (VEGF). The VEGF siRNA was conjugated to polyethylene glycol (PEG) (siRNA-PEG) before complexation with the micelles in order to improve the siRNA's stability and to prolong its retention time in the blood circulation. To further improve the in vivo biosafety, we prepared mixed micelles using mPEG-PCL together with PDMA-b-PCL copolymer. The SN-38/USPIO-loaded siRNA-PEG mixed micelleplexes passively targeted to tumor regions and synergistically facilitated VEGF silencing and chemotherapy, thus efficiently suppressing tumor growth via a multi-dose therapy regimen. Additionally, the SN-38/USPIO-loaded siRNA-PEG mixed micelleplexes acted as a negative magnetic resonance imaging (MRI) contrast agent in T2-weighted imaging, resulting in a powerful tool for the diagnosis and for tracking of the therapeutic outcomes. In summary, we established a theranostic micellar drug and gene delivery system that not only synergistically combined gene silencing and chemotherapy but also served as a negative MRI contrast agent, which reveal its potential as a novel colorectal cancer therapy.
Collapse
Affiliation(s)
- Shin-Yu Lee
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 100, Taiwan
| | - Chia-Ying Yang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 100, Taiwan
| | - Cheng-Liang Peng
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 100, Taiwan; Isotope Application Division, Institute of Nuclear Energy Research, P.O. Box 3-27, Longtan, Taoyuan, 325, Taiwan
| | - Ming-Feng Wei
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 100, Taiwan
| | - Ke-Cheng Chen
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 100, Taiwan; Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Cheng-Jung Yao
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 100, Taiwan; Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ming-Jium Shieh
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 100, Taiwan; Department of Oncology, National Taiwan University Hospital and College of Medicine, No. 7, Chung-Shan South Road, Taipei, 100, Taiwan.
| |
Collapse
|
76
|
Svenson S, Case RI, Cole RO, Hwang J, Kabir SR, Lazarus D, Lim Soo P, Ng PS, Peters C, Shum P, Sweryda-Krawiec B, Tripathi S, van der Poll D, Eliasof S. Tumor Selective Silencing Using an RNAi-Conjugated Polymeric Nanopharmaceutical. Mol Pharm 2016; 13:737-47. [DOI: 10.1021/acs.molpharmaceut.5b00608] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Sonke Svenson
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Roy I. Case
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Roderick O. Cole
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Jungyeon Hwang
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Sujan R. Kabir
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Douglas Lazarus
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Patrick Lim Soo
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Pei-Sze Ng
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Christian Peters
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Pochi Shum
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Beata Sweryda-Krawiec
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Snehlata Tripathi
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Derek van der Poll
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| | - Scott Eliasof
- Cerulean Pharma Inc., 35 Gatehouse
Drive, Waltham, Massachusetts 02451, United States
| |
Collapse
|
77
|
Rao NV, Yoon HY, Han HS, Ko H, Son S, Lee M, Lee H, Jo DG, Kang YM, Park JH. Recent developments in hyaluronic acid-based nanomedicine for targeted cancer treatment. Expert Opin Drug Deliv 2015; 13:239-52. [PMID: 26653872 DOI: 10.1517/17425247.2016.1112374] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Hyaluronic acid (HA) has emerged as a promising applicant for the tumor-targeted delivery of various therapeutic agents. Because of its biocompatibility, biodegradability and receptor-binding properties, HA has been extensively investigated as the drug delivery carrier. In this review, recent advances in HA-based nanomedicines are discussed. AREAS COVERED This review focuses on HA-based nanomedicines for the diagnosis and treatment of cancer. In particular, recent advances in HA-drug conjugates and HA-based nanoparticles for small molecular drug delivery are discussed. The bioreducible HA conjugates for small interfering ribonucleic acid delivery have been also discussed. EXPERT OPINION To develop a successful HA-based nanomedicine, it has to be prepared without significant deterioration of intrinsic property of HA. The chemical modification of HA with drugs or hydrophobic moieties may reduce the binding affinity of HA to the receptors. In addition, since the HA-based nanomedicines tend to accumulate in the liver after their systemic administration, new strategies to overcome this issue have to be developed.
Collapse
Affiliation(s)
- N Vijayakameswara Rao
- a School of Chemical Engineering , Sungkyunkwan University , Suwon , Republic of Korea
| | - Hong Yeol Yoon
- a School of Chemical Engineering , Sungkyunkwan University , Suwon , Republic of Korea
| | - Hwa Seung Han
- a School of Chemical Engineering , Sungkyunkwan University , Suwon , Republic of Korea
| | - Hyewon Ko
- b Department of Health Sciences and Technology , SAIHST, Sungkyunkwan University , Suwon , Republic of Korea
| | - Soyoung Son
- b Department of Health Sciences and Technology , SAIHST, Sungkyunkwan University , Suwon , Republic of Korea
| | - Minchang Lee
- a School of Chemical Engineering , Sungkyunkwan University , Suwon , Republic of Korea
| | - Hansang Lee
- a School of Chemical Engineering , Sungkyunkwan University , Suwon , Republic of Korea
| | - Dong-Gyu Jo
- c School of Pharmacy , Sungkyunkwan University , Suwon , Republic of Korea
| | - Young Mo Kang
- d School of Medicine , Kyungpook National University , Daegu , Republic of Korea
| | - Jae Hyung Park
- a School of Chemical Engineering , Sungkyunkwan University , Suwon , Republic of Korea.,b Department of Health Sciences and Technology , SAIHST, Sungkyunkwan University , Suwon , Republic of Korea
| |
Collapse
|
78
|
Wang T, Gantier MP, Xiang D, Bean AG, Bruce M, Zhou SF, Khasraw M, Ward A, Wang L, Wei MQ, AlShamaileh H, Chen L, She X, Lin J, Kong L, Shigdar S, Duan W. EpCAM Aptamer-mediated Survivin Silencing Sensitized Cancer Stem Cells to Doxorubicin in a Breast Cancer Model. Am J Cancer Res 2015; 5:1456-72. [PMID: 26681989 PMCID: PMC4672025 DOI: 10.7150/thno.11692] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 07/30/2015] [Indexed: 12/23/2022] Open
Abstract
Understanding the molecular basis of drug resistance and utilising this information to overcome chemoresistance remains a key challenge in oncology. Here we report that survivin, a key protein implicated in drug resistance, is overexpressed in cancer stem cell pool of doxorubicin-resistant breast cancer cells. Moreover, by utilising an active targeting system consisting of an RNA aptamer targeted against the epithelial cell adhesion molecule and a Dicer substrate survivin siRNA, we could deliver a high dose of the siRNA to cancer stem cells in xenograft tumours. Importantly, silencing of survivin with this aptamer-siRNA chimera in cancer stem cell population led to the reversal of chemoresistance, such that combined treatment with low dose of doxorubicin inhibited stemness, eliminated cancer stem cells via apoptosis, suppressed tumour growth, and prolonged survival in mice bearing chemoresistant tumours. This strategy for in vivo cancer stem cell targeting has wide application for future effective silencing of anti-death genes and in fact any dysregulated genes involved in chemoresistance and tumour relapse.
Collapse
|
79
|
Yeung BZ, Lu Z, Wientjes GM, Au JLS. High Sensitivity RT-qPCR Assay of Nonlabeled siRNA in Small Blood Volume for Pharmacokinetic Studies: Application to Survivin siRNA. AAPS JOURNAL 2015; 17:1475-82. [PMID: 26286676 DOI: 10.1208/s12248-015-9812-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/02/2015] [Indexed: 11/30/2022]
Abstract
RNAi therapeutics provide an opportunity to correct faulty genes, and several RNAi have entered clinical evaluation. The existing quantification methods typically use radioactivity- or fluorescence-labeled RNAi, require large blood volumes, and/or have a limited dynamic detection range. We established a quantitative reverse transcriptase real-time polymerase chain reaction (RT-qPCR) assay to measure RNAi; the model analyte was survivin siRNA (siSurvivin). A second siRNA was used as the internal standard. The three major steps were (a) extraction of the two siRNAs from blood or water, (b) synthesis of their cDNA by poly-A extension, and (c) qPCR of cDNA. Standard curves were established. Utility of the assay was demonstrated in a pharmacokinetic study where all 12 samples for the blood concentration-time profile were obtained from a single mouse given an intravenous dose of 1 nmole siSurvivin (prepared as lipoplex with pegylated cationic liposomes). The RT-qPCR assay was sensitive (lower detection limit of 100 fM) and had a 5 × 107-fold dynamic range and low sample volume requirement (10 μL). The 16-point standard curves constructed using whole blood samples were linear (R (2) > 0.98). The intraday and interday variations for the slopes were ≤6%, although the variations for accuracy and precision at individual concentrations were substantially higher (58-145%). Standard curves prepared with water in place of blood showed similar results (<6% difference), indicating water may be used when blood is not available. The current RT-qPCR assay enabled the measurement of nonlabeled siRNA in small volume of blood samples.
Collapse
Affiliation(s)
- Bertrand Z Yeung
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA
| | - Ze Lu
- Optimum Therapeutics LLC, 1815 Aston Ave, Suite 107, Carlsbad, California, 92008, USA
| | - Guillaume M Wientjes
- Optimum Therapeutics LLC, 1815 Aston Ave, Suite 107, Carlsbad, California, 92008, USA
| | - Jessie L-S Au
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA. .,Optimum Therapeutics LLC, 1815 Aston Ave, Suite 107, Carlsbad, California, 92008, USA. .,Taipei Medical University, Taipei, Taiwan, Republic of China.
| |
Collapse
|
80
|
Lin EW, Maynard HD. Grafting from Small Interfering Ribonucleic Acid (siRNA) as an Alternative Synthesis Route to siRNA–Polymer Conjugates. Macromolecules 2015. [DOI: 10.1021/acs.macromol.5b00846] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- En-Wei Lin
- Department of Chemistry & Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Heather D. Maynard
- Department of Chemistry & Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| |
Collapse
|
81
|
Boccellino M, Alaia C, Misso G, Cossu AM, Facchini G, Piscitelli R, Quagliuolo L, Caraglia M. Gene interference strategies as a new tool for the treatment of prostate cancer. Endocrine 2015; 49:588-605. [PMID: 26049369 DOI: 10.1007/s12020-015-0629-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/11/2015] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PCa) is one of the most common cancer in men. It affects older men and the incidence increases with age; the median age at diagnosis is 67 years. The diagnosis of PCa is essentially based on three tools: digital rectal exam, serum concentration of prostate specific antigen, and transrectal ultrasound-guided biopsy. Currently, the therapeutic treatments of this cancer are different and range from the prostatectomy to hormonal therapy, to radiation therapy, to immunotherapy, and to chemotherapy. However, additional efforts are required in order to find new weapons for the treatment of metastatic setting of disease. The purpose of this review is to highlight new therapeutic strategies based on gene interference; in fact, numerous siRNA and miRNA in the therapeutic treatment of PCa are reported below.
Collapse
Affiliation(s)
- Mariarosaria Boccellino
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via L. De Crecchio, 7, 80138, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Yang L, Li W, Huang Y, Zhou Y, Chen T. Rational Design of Cancer-Targeted Benzoselenadiazole by RGD Peptide Functionalization for Cancer Theranostics. Macromol Rapid Commun 2015. [DOI: 10.1002/marc.201500243] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Liye Yang
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Wenying Li
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Yanyu Huang
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Yangliang Zhou
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Tianfeng Chen
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| |
Collapse
|
83
|
Li Y, Yang J, Xu B, Gao F, Wang W, Liu W. Enhanced Therapeutic siRNA to Tumor Cells by a pH-Sensitive Agmatine-Chitosan Bioconjugate. ACS APPLIED MATERIALS & INTERFACES 2015; 7:8114-24. [PMID: 25832629 DOI: 10.1021/acsami.5b00851] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Charge-conversional naturally occurring chitosan-agmatine bioconjugates are prepared by dimethylmaleic anhydride (DMA) modification and the nucleophilic reaction between tosyl of tosylated chitosan and primary amine of agmatine. These bioconjugates (CS-DM-Agm) are shown to condense siRNA into nanocomplexes, which are stable in the presence of serum at physical pH values. Furthermore, the surface charge of complexes can tune from negative to positive while pH is changed to weak acid tumor micromilieu, thus facilitating the target cancer cell internalization in resisting serum adsorption. More importantly, this smart biogenic system shows remarkable gene silencing efficiency and a high apoptotic rate of tumor cells both in vitro and in vivo, indicating its great potential for cancer therapy.
Collapse
Affiliation(s)
- Yongmao Li
- †School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, P. R. China
- ‡Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
| | - Jianhai Yang
- †School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, P. R. China
| | - Bing Xu
- †School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, P. R. China
| | - Fei Gao
- †School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, P. R. China
| | - Wei Wang
- †School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, P. R. China
- §State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, P. R. China
| | - Wenguang Liu
- †School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, P. R. China
- ‡Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
| |
Collapse
|
84
|
Polylysine-modified polyethylenimines as siRNA carriers for effective tumor treatment. CHINESE JOURNAL OF POLYMER SCIENCE 2015. [DOI: 10.1007/s10118-015-1632-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
85
|
Hörtz C, Birke A, Kaps L, Decker S, Wächtersbach E, Fischer K, Schuppan D, Barz M, Schmidt M. Cylindrical Brush Polymers with Polysarcosine Side Chains: A Novel Biocompatible Carrier for Biomedical Applications. Macromolecules 2015. [DOI: 10.1021/ma502497x] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Christian Hörtz
- Institute
for Physical Chemistry, Johannes Gutenberg University, Welder Weg
11, D-55099 Mainz, Germany
| | - Alexander Birke
- Institute
for Organic Chemistry, Johannes Gutenberg University, Duesbergweg
10-14, D-55099 Mainz, Germany
| | - Leonard Kaps
- Institute
of Translational Immunology and Research Center for Immunotherapy,
University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse
1, D-55131 Mainz, Germany
| | - Sandra Decker
- Institute
for Physical Chemistry, Johannes Gutenberg University, Welder Weg
11, D-55099 Mainz, Germany
| | - Eva Wächtersbach
- Institute
for Physical Chemistry, Johannes Gutenberg University, Welder Weg
11, D-55099 Mainz, Germany
| | - Karl Fischer
- Institute
for Physical Chemistry, Johannes Gutenberg University, Welder Weg
11, D-55099 Mainz, Germany
| | - Detlef Schuppan
- Institute
of Translational Immunology and Research Center for Immunotherapy,
University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse
1, D-55131 Mainz, Germany
| | - Matthias Barz
- Institute
for Organic Chemistry, Johannes Gutenberg University, Duesbergweg
10-14, D-55099 Mainz, Germany
| | - Manfred Schmidt
- Institute
for Physical Chemistry, Johannes Gutenberg University, Welder Weg
11, D-55099 Mainz, Germany
| |
Collapse
|
86
|
Monaghan M, Greiser U, Cao H, Wang W, Pandit A. An antibody fragment functionalized dendritic PEGylated poly(2-(dimethylamino)ethyl diacrylate) as a vehicle of exogenous microRNA. Drug Deliv Transl Res 2015; 2:406-14. [PMID: 25787178 DOI: 10.1007/s13346-012-0097-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The translation of interfering RNA to the clinic requires more effective delivery agents to enable safe and efficient delivery. The aim of this work was to create a multi-functional delivery agent using deactivation enhanced ATRP synthesis of poly(dimethylamino)ethyl methacrylate (pDMAEMA)-co-PEGMEA/PEGDA (pD-b-P/DA) with linear pDMAEMA as a macro-initiator. The pD-b-P/DA was characterized for its potential to bind synthetic microRNA mimics to form structures and reacted with antibody-derived fragments (Fabs) using Michael-type addition. Conjugation of antibody fragments was verified using SDS-PAGE. Functional delivery of these interfering RNA complexes was proven using a dual luciferase reporter assay. Functional silencing of a reporter gene was improved by complexation of microRNA mimics with pD-b-P/DA alone and with Fab-decorated pD-b-P/DA. The improved silencing with Fab-decorated pD-b-P/DA was evident at 48 h but disappeared at 96 h. The resultant agent enables complexation of nucleic acid (microRNA mimic) and facile conjugation of antibody fragments via a Michael-type addition. In conclusion, this platform is effective at silencing in this reporter system and has potential as an effective delivery system of interfering RNA.
Collapse
Affiliation(s)
- M Monaghan
- Network of Excellence for Functional Biomaterials, National University of Ireland, Galway, Ireland
| | | | | | | | | |
Collapse
|
87
|
Wu X, Yamamoto H, Nakanishi H, Yamamoto Y, Inoue A, Tei M, Hirose H, Uemura M, Nishimura J, Hata T, Takemasa I, Mizushima T, Hossain S, Akaike T, Matsuura N, Doki Y, Mori M. Innovative delivery of siRNA to solid tumors by super carbonate apatite. PLoS One 2015; 10:e0116022. [PMID: 25738937 PMCID: PMC4349808 DOI: 10.1371/journal.pone.0116022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 11/19/2014] [Indexed: 01/19/2023] Open
Abstract
RNA interference (RNAi) technology is currently being tested in clinical trials for a limited number of diseases. However, systemic delivery of small interfering RNA (siRNA) to solid tumors has not yet been achieved in clinics. Here, we introduce an in vivo pH-sensitive delivery system for siRNA using super carbonate apatite (sCA) nanoparticles, which is the smallest class of nanocarrier. These carriers consist simply of inorganic ions and accumulate specifically in tumors, yet they cause no serious adverse events in mice and monkeys. Intravenously administered sCA-siRNA abundantly accumulated in the cytoplasm of tumor cells at 4 h, indicating quick achievement of endosomal escape. sCA-survivin-siRNA induced apoptosis in HT29 tumors and significantly inhibited in vivo tumor growth of HCT116, to a greater extent than two other in vivo delivery reagents. With innovative in vivo delivery efficiency, sCA could be a useful nanoparticle for the therapy of solid tumors.
Collapse
Affiliation(s)
- Xin Wu
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Hirofumi Yamamoto
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
- * E-mail:
| | | | - Yuki Yamamoto
- Nakanishi Gastroenterological Research Institute, Sakai, Japan
| | - Akira Inoue
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Mitsuyoshi Tei
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hajime Hirose
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Mamoru Uemura
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Junichi Nishimura
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Taishi Hata
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Ichiro Takemasa
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tsunekazu Mizushima
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Sharif Hossain
- Department of Biomolecular Engineering, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
- Biomaterials Center for Regenerative Medical Engineering, Foundation for Advancement of International Science, Tsukuba, Japan
| | - Toshihiro Akaike
- Department of Biomolecular Engineering, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
- Biomaterials Center for Regenerative Medical Engineering, Foundation for Advancement of International Science, Tsukuba, Japan
| | - Nariaki Matsuura
- Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Yuichiro Doki
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Masaki Mori
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
88
|
Essex S, Navarro G, Sabhachandani P, Chordia A, Trivedi M, Movassaghian S, Torchilin VP. Phospholipid-modified PEI-based nanocarriers for in vivo siRNA therapeutics against multidrug-resistant tumors. Gene Ther 2015; 22:257-266. [PMID: 25354685 PMCID: PMC4352110 DOI: 10.1038/gt.2014.97] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 09/05/2014] [Accepted: 09/17/2014] [Indexed: 12/17/2022]
Abstract
Multidrug resistance (MDR) mediated by P-glycoprotein overexpression in solid tumors is a major factor in the failure of many forms of chemotherapy. Here we evaluated phospholipid-modified, low-molecular-weight polyethylenimine (DOPE-PEI) nanocarriers for intravenous delivery of anti-P-pg siRNA to tumors with the final goal of modulating MDR in breast cancer. First, we studied the biodistribution of DOPE-PEI nanocarriers and the effect of PEG coating in a subcutaneous breast tumor model. Four hours postinjection, PEGylated carriers showed an 8% injected dose (ID) accumulation in solid tumor via the enhanced permeability and retention effect and 22% ID in serum due to a prolonged, PEG-mediated circulation. Second, we established the therapeutic efficacy and safety of DOPE-PEI/siRNA-mediated P-gp downregulation in combination with doxorubicin (Dox) chemotherapy in MCF-7/MDR xenografts. Weekly injection of siRNA nanopreparations and Dox for up to 5 weeks sensitized the tumors to otherwise non-effective doses of Dox and decreased the tumor volume by threefold vs controls. This therapeutic improvement in response to Dox was attributed to the significant, sequence-specific P-gp downregulation in excised tumors mediated by the DOPE-PEI formulations.
Collapse
Affiliation(s)
- Sean Essex
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Gemma Navarro
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Pooja Sabhachandani
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Aabha Chordia
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Malav Trivedi
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Sara Movassaghian
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Vladimir P. Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
89
|
Han L, Tang C, Yin C. Enhanced antitumor efficacies of multifunctional nanocomplexes through knocking down the barriers for siRNA delivery. Biomaterials 2015; 44:111-21. [PMID: 25617131 DOI: 10.1016/j.biomaterials.2014.12.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 12/10/2014] [Accepted: 12/20/2014] [Indexed: 02/07/2023]
Abstract
Multifunctional nanocomplexes (NCs) consisting of urocanic acid-modified galactosylated trimethyl chitosan (UA-GT) conjugates as polymeric vectors, poly(allylamine hydrochloride)-citraconic anhydride (PAH-Cit) as charge-reversible crosslinkers, and vascular endothelial growth factor (VEGF) siRNA as therapeutic genes, were rationally designed to simultaneously overcome the extracellular, cellular, and intracellular barriers for siRNA delivery. The strong physical stability of UA-GT/PAH-Cit/siRNA NCs (UA-GT NCs) at pH 7.4 and 6.5 endowed protection from massive dilution, competitive ions, and ubiquitous nucleases in the blood and tumorous microenvironment. Their internalization into hepato-carcinoma cells was facilitated through the recognition of galactose receptors, followed by effective escape from endosomes/lysosomes owing to the strong buffering capacity of imidazole residues. At the meantime, the endosomal/lysosomal acidity triggered the charge reversal of PAH-Cit in UA-GT NCs, thus evoking their structural disassembly and subsequently accelerated release of siRNA in the cytosol. As a result, robust in vivo performance in terms of both gene silencing and tumor inhibition was achieved by UA-GT NCs at a low siRNA dose. Moreover, neither histological nor hematological toxicity was detected following repeated intravenous administration. Therefore, UA-GT NCs potentially served as an efficient and safe candidate in the treatment of hepatocellular carcinoma through knocking down the overall barriers for siRNA delivery.
Collapse
Affiliation(s)
- Lu Han
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Cui Tang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200433, China.
| |
Collapse
|
90
|
Ren X, Feng Y, Guo J, Wang H, Li Q, Yang J, Hao X, Lv J, Ma N, Li W. Surface modification and endothelialization of biomaterials as potential scaffolds for vascular tissue engineering applications. Chem Soc Rev 2015; 44:5680-742. [DOI: 10.1039/c4cs00483c] [Citation(s) in RCA: 359] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review highlights the recent developments of surface modification and endothelialization of biomaterials in vascular tissue engineering applications.
Collapse
Affiliation(s)
- Xiangkui Ren
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Yakai Feng
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Jintang Guo
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Haixia Wang
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Qian Li
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Jing Yang
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Xuefang Hao
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Juan Lv
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Nan Ma
- Institute of Chemistry and Biochemistry
- Free University of Berlin
- D-14195 Berlin
- Germany
| | - Wenzhong Li
- Department of Cardiac Surgery
- University of Rostock
- D-18057 Rostock
- Germany
| |
Collapse
|
91
|
Yang H, Li Y, Li T, Xu M, Chen Y, Wu C, Dang X, Liu Y. Multifunctional core/shell nanoparticles cross-linked polyetherimide-folic acid as efficient Notch-1 siRNA carrier for targeted killing of breast cancer. Sci Rep 2014; 4:7072. [PMID: 25400232 PMCID: PMC4233336 DOI: 10.1038/srep07072] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/29/2014] [Indexed: 12/13/2022] Open
Abstract
In gene therapy, how genetic therapeutics can be efficiently and safely delivered into target tissues/cells remains a major obstacle to overcome. To address this issue, nanoparticles consisting of non-covalently coupled polyethyleneimine (PEI) and folic acid (FA) to the magnetic and fluorescent core/shell of Fe3O4@SiO2(FITC) was tested for their ability to deliver Notch-1 shRNA. Our results showed that Fe3O4@SiO2(FITC)/PEI-FA/Notch-1 shRNA nanoparticles are 64 nm in diameter with well dispersed and superparamagnetic. These nanoparticles with on significant cytotoxicity are capable of delivering Notch-1 shRNA into human breast cancer MDA-MB-231 cells with high efficiency while effectively protected shRNA from degradation by exogenous DNaseI and nucleases. Magnetic resonance (MR) imaging and fluorescence microscopy showed significant preferential uptake of Fe3O4@SiO2(FITC)/PEI-FA/Notch-1 shRNA nanocomplex by MDA-MB-231 cells. Transfected MDA-MB-231 cells exhibited significantly decreased expression of Notch-1, inhibited cell proliferation, and increased cell apoptosis, leading to the killing of MDA-MB-231 cells. In light of the magnetic targeting capabilities of Fe3O4@SiO2(FITC)/PEI-FA, our results show that by complexing with a second molecular targeting therapeutic, such as Notch-1 shRNA in this report, Fe3O4@SiO2(FITC)/PEI-FA can be exploited as a novel, non-viral, and concurrent targeting delivery system for targeted gene therapy as well as for MR imaging in cancer diagnosis.
Collapse
Affiliation(s)
- Hong Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Ying Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Tingting Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Min Xu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Yin Chen
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Xitong Dang
- Division of Trauma, Surgical Critical Care and Burns, University of California San Diego, CA 92103, USA
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| |
Collapse
|
92
|
Borna H, Imani S, Iman M, Azimzadeh Jamalkandi S. Therapeutic face of RNAi: in vivo challenges. Expert Opin Biol Ther 2014; 15:269-85. [PMID: 25399911 DOI: 10.1517/14712598.2015.983070] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION RNA interference is a sequence-specific gene silencing phenomenon in which small interfering RNAs (siRNAs) can trigger gene transcriptional and post-transcriptional silencing. This phenomenon represents an emerging therapeutic approach for in vivo studies by efficient delivery of specific synthetic siRNAs against diseases. Therefore, simultaneous development of synthetic siRNAs along with novel delivery techniques is considered as novel and interesting therapeutic challenges. AREAS COVERED This review provides a basic explanation to siRNA signaling pathways and their therapeutic challenges. Here, we provide a comprehensive explanation to failed and successful trials and their in vivo challenges. EXPERT OPINION Specific, efficient and targeted delivery of siRNAs is the major concern for their in vivo administrations. Also, anatomical barriers, drug stability and availability, immunoreactivity and existence of various delivery routes, different genetic backgrounds are major clinical challenges. However, successful administration of siRNA-based drugs is expected during foreseeable features. But, their systemic applications will depend on strong targeted drug delivery strategies.
Collapse
Affiliation(s)
- Hojat Borna
- Baqiyatallah University of Medical Sciences, Chemical Injuries Research Center , Tehran , Iran
| | | | | | | |
Collapse
|
93
|
Hong CA, Nam YS. Functional nanostructures for effective delivery of small interfering RNA therapeutics. Am J Cancer Res 2014; 4:1211-32. [PMID: 25285170 PMCID: PMC4183999 DOI: 10.7150/thno.8491] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 02/23/2014] [Indexed: 02/04/2023] Open
Abstract
Small interfering RNA (siRNA) has proved to be a powerful tool for target-specific gene silencing via RNA interference (RNAi). Its ability to control targeted gene expression gives new hope to gene therapy as a treatment for cancers and genetic diseases. However, siRNA shows poor pharmacological properties, such as low serum stability, off-targeting, and innate immune responses, which present a significant challenge for clinical applications. In addition, siRNA cannot cross the cell membrane for RNAi activity because of its anionic property and stiff structure. Therefore, the development of a safe, stable, and efficient system for the delivery of siRNA therapeutics into the cytoplasm of targeted cells is crucial. Several nanoparticle platforms for siRNA delivery have been developed to overcome the major hurdles facing the therapeutic uses of siRNA. This review covers a broad spectrum of non-viral siRNA delivery systems developed for enhanced cellular uptake and targeted gene silencing in vitro and in vivo and discusses their characteristics and opportunities for clinical applications of therapeutic siRNA.
Collapse
|
94
|
Nanocarrier mediated delivery of siRNA/miRNA in combination with chemotherapeutic agents for cancer therapy: current progress and advances. J Control Release 2014; 194:238-56. [PMID: 25204288 DOI: 10.1016/j.jconrel.2014.09.001] [Citation(s) in RCA: 262] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/29/2014] [Accepted: 09/01/2014] [Indexed: 12/21/2022]
Abstract
Chemotherapeutic agents have certain limitations when it comes to treating cancer, the most important being severe side effects along with multidrug resistance developed against them. Tumor cells exhibit drug resistance due to activation of various cellular level processes viz. activation of drug efflux pumps, anti-apoptotic defense mechanisms, etc. Currently, RNA interference (RNAi) based therapeutic approaches are under vibrant scrutinization to seek cancer cure. Especially small interfering RNA (siRNA) and micro RNA (miRNA), are able to knock down the carcinogenic genes by targeting the mRNA expression, which underlies the uniqueness of this therapeutic approach. Recent research focus in the regime of cancer therapy involves the engagement of targeted delivery of siRNA/miRNA in combinations with other therapeutic agents (such as gene, DNA or chemotherapeutic drug) for targeting permeability glycoprotein (P-gp), multidrug resistant protein 1 (MRP-1), B-cell lymphoma (BCL-2) and other targets that are mainly responsible for resistance in cancer therapy. RNAi-chemotherapeutic drug combinations have also been found to be effective against different molecular targets as well and can increase the sensitization of cancer cells to therapy several folds. However, due to stability issues associated with siRNA/miRNA suitable protective carrier is needed and nanotechnology based approaches have been widely explored to overcome these drawbacks. Furthermore, it has been univocally advocated that the co-delivery of siRNA/miRNA with other chemodrugs significantly enhances their capability to overcome cancer resistance compared to naked counterparts. The objective of this article is to review recent nanocarrier based approaches adopted for the delivery of siRNA/miRNA combinations with other anticancer agents (siRNA/miRNA/pDNA/chemodrugs) to treat cancer.
Collapse
|
95
|
Ballarín-González B, Ebbesen MF, Howard KA. Polycation-based nanoparticles for RNAi-mediated cancer treatment. Cancer Lett 2014; 352:66-80. [DOI: 10.1016/j.canlet.2013.09.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/16/2013] [Accepted: 09/18/2013] [Indexed: 12/19/2022]
|
96
|
Dawidczyk CM, Russell LM, Searson PC. Nanomedicines for cancer therapy: state-of-the-art and limitations to pre-clinical studies that hinder future developments. Front Chem 2014; 2:69. [PMID: 25202689 PMCID: PMC4142601 DOI: 10.3389/fchem.2014.00069] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/05/2014] [Indexed: 01/31/2023] Open
Abstract
The ability to efficiently deliver a drug or gene to a tumor site is dependent on a wide range of factors including circulation time, interactions with the mononuclear phagocyte system, extravasation from circulation at the tumor site, targeting strategy, release from the delivery vehicle, and uptake in cancer cells. Nanotechnology provides the possibility of creating delivery systems where the design constraints are decoupled, allowing new approaches for reducing the unwanted side effects of systemic delivery, increasing tumor accumulation, and improving efficacy. The physico-chemical properties of nanoparticle-based delivery platforms introduce additional complexity associated with pharmacokinetics, tumor accumulation, and biodistribution. To assess the impact of nanoparticle-based delivery systems, we first review the design strategies and pharmacokinetics of FDA-approved nanomedicines. Next we review nanomedicines under development, summarizing the range of nanoparticle platforms, strategies for targeting, and pharmacokinetics. We show how the lack of uniformity in preclinical trials prevents systematic comparison and hence limits advances in the field.
Collapse
Affiliation(s)
- Charlene M Dawidczyk
- Institute for Nanobiotechnology, Johns Hopkins University Baltimore, MD, USA ; Johns Hopkins Center of Cancer Nanotechnology Excellence, Johns Hopkins University Baltimore, MD, USA ; Department of Materials Science and Engineering, Johns Hopkins University Baltimore, MD, USA
| | - Luisa M Russell
- Institute for Nanobiotechnology, Johns Hopkins University Baltimore, MD, USA ; Johns Hopkins Center of Cancer Nanotechnology Excellence, Johns Hopkins University Baltimore, MD, USA ; Department of Materials Science and Engineering, Johns Hopkins University Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University Baltimore, MD, USA ; Johns Hopkins Center of Cancer Nanotechnology Excellence, Johns Hopkins University Baltimore, MD, USA ; Department of Materials Science and Engineering, Johns Hopkins University Baltimore, MD, USA
| |
Collapse
|
97
|
Ku SH, Kim K, Choi K, Kim SH, Kwon IC. Tumor-targeting multifunctional nanoparticles for siRNA delivery: recent advances in cancer therapy. Adv Healthc Mater 2014; 3:1182-93. [PMID: 24577795 DOI: 10.1002/adhm.201300607] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/20/2014] [Indexed: 11/06/2022]
Abstract
RNA interference (RNAi) is a naturally occurring regulatory process that controls posttranscriptional gene expression. Small interfering RNA (siRNA), a common form of RNAi-based therapeutics, offers new opportunities for cancer therapy via silencing specific genes, which are associated to cancer progress. However, clinical applications of RNAi-based therapy are still limited due to the easy degradation of siRNA during body circulation and the difficulty in the delivery of siRNA to desired tissues and cells. Thus, there have been many efforts to develop efficient siRNA delivery systems, which protect siRNA from serum nucleases and deliver siRNA to the intracellular region of target cells. Here, the recent advances in siRNA nanocarriers, which possess tumor-targeting ability are reviewed; various nanoparticle systems and their antitumor effects are summarized. The development of multifunctional nanocarriers for theranostics or combinatorial therapy is also discussed.
Collapse
Affiliation(s)
- Sook Hee Ku
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology (KIST); Seoul 136-791 Republic of Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology (KIST); Seoul 136-791 Republic of Korea
| | - Kuiwon Choi
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology (KIST); Seoul 136-791 Republic of Korea
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology (KIST); Seoul 136-791 Republic of Korea
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology (KIST); Seoul 136-791 Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology; Korea University; Seoul 136-701 Republic of Korea
| |
Collapse
|
98
|
Direct cytosolic siRNA delivery by reconstituted high density lipoprotein for target-specific therapy of tumor angiogenesis. Biomaterials 2014; 35:7214-27. [DOI: 10.1016/j.biomaterials.2014.05.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 05/02/2014] [Indexed: 12/16/2022]
|
99
|
Shima F, Shudo M, Akagi T, Akashi M. Preparation of siRNA Carrier Based on Boronic Acid-functionalized Amphiphilic Poly(γ-glutamic acid) Nanoparticles. CHEM LETT 2014. [DOI: 10.1246/cl.140060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Fumiaki Shima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University
| | - Manami Shudo
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University
| | - Takami Akagi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University
| | - Mitsuru Akashi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University
| |
Collapse
|
100
|
Chen S, Feng J, Ma L, Liu Z, Yuan W. RNA interference technology for anti-VEGF treatment. Expert Opin Drug Deliv 2014; 11:1471-80. [DOI: 10.1517/17425247.2014.926886] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|