51
|
Ben David-Naim M, Grad E, Aizik G, Nordling-David MM, Moshel O, Granot Z, Golomb G. Polymeric nanoparticles of siRNA prepared by a double-emulsion solvent-diffusion technique: Physicochemical properties, toxicity, biodistribution and efficacy in a mammary carcinoma mice model. Biomaterials 2017; 145:154-167. [PMID: 28863309 DOI: 10.1016/j.biomaterials.2017.08.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 01/06/2023]
Abstract
siRNA-loaded nanoparticles (NPs) administered systemically can overcome the poor stability and rapid elimination of free double-stranded RNA in circulation, resulting in increased tumor accumulation and efficacy. siRNA against osteopontin (siOPN), a protein involved in breast cancer development, was encapsulated in poly(D,L-lactic-co-glycolic acid) NPs by a double emulsion solvent diffusion (DESD) technique. We also compared the effect of polyethylenimine (PEI) molecular weight (800 Da and 25 kDa), used as the counter-ion for siRNA complexation, on the physicochemical properties of the NPs, cytotoxicity, and cellular uptake. NPs prepared by the DESD technique were obtained at the desired size (∼170 nm) using both types of PEIs, and were characterized with a neutral surface charge, high encapsulation yield (up to ∼60%), siOPN concentration of 5.6-8.4 μg/mg, stability in physiologic conditions in vitro and in vivo, and long-term shelf-life stability (> 3 years). The NPs prepared using both PEIs exhibited no cytotoxicity in primary smooth muscle culture, and no detrimental effect on mice liver enzymes following their IV administration. Following cellular uptake and biodistribution studies, the therapeutic potential of the NPs was demonstrated by a significant decrease of tumor progression and size in an ectopic xenograft model of mammary carcinoma in mice.
Collapse
Affiliation(s)
- Meital Ben David-Naim
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Etty Grad
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gil Aizik
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mirjam M Nordling-David
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofra Moshel
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Zvi Granot
- Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gershon Golomb
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
52
|
Khalid A, Persano S, Shen H, Zhao Y, Blanco E, Ferrari M, Wolfram J. Strategies for improving drug delivery: nanocarriers and microenvironmental priming. Expert Opin Drug Deliv 2017; 14:865-877. [PMID: 27690153 PMCID: PMC5584706 DOI: 10.1080/17425247.2017.1243527] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The ultimate goal in the field of drug delivery is to exclusively direct therapeutic agents to pathological tissues in order to increase therapeutic efficacy and eliminate side effects. This goal is challenging due to multiple transport obstacles in the body. Strategies that improve drug transport exploit differences in the characteristics of normal and pathological tissues. Within the field of oncology, these concepts have laid the groundwork for a new discipline termed transport oncophysics. Areas covered: Efforts to improve drug biodistribution have mainly focused on nanocarriers that enable preferential accumulation of drugs in diseased tissues. A less common approach to enhance drug transport involves priming strategies that modulate the biological environment in ways that favor localized drug delivery. This review discusses a variety of priming and nanoparticle design strategies that have been used for drug delivery. Expert opinion: Combinations of priming agents and nanocarriers are likely to yield optimal drug distribution profiles. Although priming strategies have yet to be widely implemented, they represent promising solutions for overcoming biological transport barriers. In fact, such strategies are not restricted to priming the tumor microenvironment but can also be directed toward healthy tissue in order to reduce nanoparticle uptake.
Collapse
Affiliation(s)
- Ayesha Khalid
- Medical Program, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Stefano Persano
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
- Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
53
|
Tumor-selective lipopolyplex encapsulated small active RNA hampers colorectal cancer growth in vitro and in orthotopic murine. Biomaterials 2017; 141:13-28. [PMID: 28666099 DOI: 10.1016/j.biomaterials.2017.06.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 06/21/2017] [Accepted: 06/21/2017] [Indexed: 02/06/2023]
Abstract
Small active RNA (saRNA)-induced gene activation (RNAa) is a novel strategy to treat cancer. Our previous work proved that the p21-saRNA-322 successfully hindered colorectal cancer growth by activating p21 gene. However, the barrier for successful saRNA therapy is lack of efficient drug delivery. In the present study, a rectal delivery system entitled p21-saRNA-322 encapsulated tumor-selective lipopolyplex (TSLPP-p21-saRNA-322) which consist of PEI/p21-saRNA-322 polyplex core and hyaluronan (HA) modulated lipid shell was developed to treat colorectal cancer. Our results showed that this system maintained at the rectum for more than 6 h and preferentially accumulated at tumor site. CD44 knock down experiment instructed that the superb cellular uptake of TSLPP-p21-saRNA-322 attributed to HA-CD44 recognition. An orthotopic model of bio-luminescence human colorectal cancer in mice was developed using microsurgery and TSLPP-p21-saRNA-322 demonstrated a superior antitumor efficacy in vitro and in vivo. Our results provide preclinical proof-of-concept for a novel method to treat colorectal cancer by rectal administration of TSLPP formulated p21-saRNA-322.
Collapse
|
54
|
Wu T, Wang L, Ding S, You Y. Fluorinated PEG-Polypeptide Polyplex Micelles Have Good Serum-Resistance and Low Cytotoxicity for Gene Delivery. Macromol Biosci 2017; 17. [PMID: 28524376 DOI: 10.1002/mabi.201700114] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Indexed: 11/09/2022]
Abstract
A novel PEGylation polypeptide, poly(ethylene glycol)-b-poly(l-lysine)-b-poly(l-cysteine) (PEG-PLL-PCys) triblock copolymer is synthesized via the sequential ring-opening polymerization of amino acid N-carboxyanhydrides initiated by methoxypolyethylene glycol amine (mPEG-NH2 , Mw is 2 kDa). Subsequently, the obtained polypeptide is partially conjugated with fluorocarbon chains via disulfide exchange reaction. PLL segment can condense plasmid DNA through an electrostatic force to form a complex core, PEG segment surrounding the complex like a corona can prevent the complex from precipitation and reduce the adsorption of serum, while PCys segment with fluorocarbon can enhance the cellular uptake and the stability of the formed polyplex micelles in physiological conditions. Experiment results exhibit that the fluorinated polypeptides have low cytotoxicity and good gene transfection efficiency even in the presence of 50% fetal bovine serum.
Collapse
Affiliation(s)
- Ting Wu
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Longhai Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Shenggang Ding
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Yezi You
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
55
|
Zhang T, Guo W, Zhang C, Yu J, Xu J, Li S, Tian JH, Wang PC, Xing JF, Liang XJ. Transferrin-Dressed Virus-like Ternary Nanoparticles with Aggregation-Induced Emission for Targeted Delivery and Rapid Cytosolic Release of siRNA. ACS APPLIED MATERIALS & INTERFACES 2017; 9:16006-16014. [PMID: 28447465 PMCID: PMC5545884 DOI: 10.1021/acsami.7b03402] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Viruses have evolved to be outstandingly efficient at gene delivery, but their use as vectors is limited by safety risks. Inspired by the structure of viruses, we constructed a virus-mimicking vector (denoted as TR4@siRNA@Tf NCs) with virus-like architecture and infection properties. Composed of a hydrophilic peptide, an aggregation-induced emission (AIE) luminogen, and a lipophilic tail, TR4 imitates the viral capsid and endows the vector with AIE properties as well as efficient siRNA compaction. The outer glycoprotein transferrin (Tf) mimics the viral envelope protein and endows the vector with reduced cytotoxicity as well as enhanced targeting capability. Because of the strong interaction between Tf and transferrin receptors on the cell surface, the Tf coating can accelerate the intracellular release of siRNA into the cytosol. Tf and TR4 are eventually cycled back to the cell membrane. Our results confirmed that the constructed siRNA@TR4@Tf NCs show a high siRNA silencing efficiency of 85% with significantly reduced cytotoxicity. These NCs have comparable transfection ability to natural viruses while avoiding the toxicity issues associated with typical nonviral vectors. Therefore, this proposed virus-like siRNA vector, which integrates the advantages of both viral and nonviral vectors, should find many potential applications in gene therapy.
Collapse
Affiliation(s)
- Tingbin Zhang
- School of Chemical Engineering and Technology, Tianjin University, No. 135 Yaguan Road, Haihe Education Park, Jinnan District, Tianjin 300350, China
| | - Weisheng Guo
- CAS Center for Excellence in Nanoscience, Chinese Academy of Sciences, CAS Key Laboratory for Biological Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunqiu Zhang
- CAS Center for Excellence in Nanoscience, Chinese Academy of Sciences, CAS Key Laboratory for Biological Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Yu
- College of Materials Science and Engineering, Zhejiang University of Technology, No. 18 Chaowang Road, Hangzhou 310014, China
| | - Jing Xu
- CAS Center for Excellence in Nanoscience, Chinese Academy of Sciences, CAS Key Laboratory for Biological Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyi Li
- CAS Center for Excellence in Nanoscience, Chinese Academy of Sciences, CAS Key Laboratory for Biological Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian-Hua Tian
- School of Chemical Engineering and Technology, Tianjin University, No. 135 Yaguan Road, Haihe Education Park, Jinnan District, Tianjin 300350, China
| | - Paul C. Wang
- Laboratory of Molecular Imaging, Department of Radiology, Howard University, Washington, D.C. 20060, United States
- College of Science and Engineering, Fu Jen Catholic University, Taipei 24205, Taiwan
| | - Jin-Feng Xing
- School of Chemical Engineering and Technology, Tianjin University, No. 135 Yaguan Road, Haihe Education Park, Jinnan District, Tianjin 300350, China
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, Chinese Academy of Sciences, CAS Key Laboratory for Biological Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
56
|
Disassembly of micelle-like polyethylenimine nanocomplexes for siRNA delivery: High transfection efficiency and reduced toxicity achieved by simple reducible lipid modification. J Colloid Interface Sci 2017; 504:633-644. [PMID: 28618383 DOI: 10.1016/j.jcis.2017.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/30/2017] [Accepted: 05/03/2017] [Indexed: 12/18/2022]
Abstract
Amphiphilic compounds consisting of polycations and lipid segments are well established as building blocks for the construction of siRNA carriers. They are capable of forming nanoparticles with high-affinity positive charges for siRNA in aqueous media due to their intra- and/or intermolecular hydrophobic and electrostatic interactions. Unfortunately, safety and efficiency of lipid-modified polycations as the two great challenges to the clinical application need to be improved. Beyond that, the role of the hydrophobic segment in the process of siRNA delivery is elusive. Herein, in this study, branched polyethylenimine with a molecular weight of 600 (bPEI600) was grafted with reducible lipids via Michael addition reaction between amines and alkyl acrylates. Reducible amphiphilic polyethylenimines (PEIs) were able to condense siRNA into nanoparticles and disassemble under the reductive environment. Investigations with these materials in vitro revealed that the polymers with higher grafting degree provided high luciferase knockdown efficacies even at lower N/P ratios and the polymers with longer lipid chain displayed greater cellular uptake rate. Interestingly, the polymers with lower grafting degree had efficient cellular uptake than native bPEI600, although their in luciferase knockdown assays were most likely inefficient. The inconsistency between the cellular uptake profile and silencing efficacy proved that the intracellular trafficking of siRNA was a bottleneck for siRNA delivery with some polymers prepared in this study. As expected, reducible lipid-modified PEIs were equally efficient and much less toxic compared to non-reducible counterparts and might provide broader therapeutic windows. These findings showed the feasibility of reducible lipid-modified PEIs as carriers for therapeutic siRNA.
Collapse
|
57
|
Minardi S, Pandolfi L, Taraballi F, Wang X, De Rosa E, Mills ZD, Liu X, Ferrari M, Tasciotti E. Enhancing Vascularization through the Controlled Release of Platelet-Derived Growth Factor-BB. ACS APPLIED MATERIALS & INTERFACES 2017; 9:14566-14575. [PMID: 28393518 DOI: 10.1021/acsami.6b13760] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Using delivery systems to control the in vivo release of growth factors (GFs) for tissue engineering applications is extremely desirable as the clinical use of GFs is limited by their fast in vivo turnover. Hence, the development of effective platforms that are able to finely control the release of GFs in vivo remains a challenge. Herein, we investigated the ability of multiscale microspheres, composed by a nanostructured silicon multistage vector (MSV) core and a poly(dl-lactide-co-glycolide) acid (PLGA) forming outer shell (PLGA-MSV), to release functional platelet-derived growth factor-BB (PDGF-BB) to induce in vivo localized neovascularization. The in vitro release of PDGF-BB was assessed by enzyme-linked immunosorbent assay (ELISA) over 2 weeks and showed a sustained, zero-order release kinetics. The ability to promote in vivo localized neovascularization was investigated in a subcutaneous injection model in BALB/c mice and followed by intravital microscopy up to 2 weeks. Fully functional newly formed vessels were found within the area where PLGA-MSVs were localized and covered 3.0 ± 0.9 and 19 ± 5.1% at 7 and 14 days, respectively, showing a 6-fold increase in 1 week. The distribution of CD31+ and α-SMA+ cells was detected by immunofluorescence on harvested tissues. CD31 was significantly more expressed (4-fold increase) compared to the untreated control. Finally, the level of up-regulation of angiogenesis-associated genes (Vegfa, Vwf, and Col3a1) was assessed by q-PCR, resulting in a significantly higher expression where PLGA-MSVs were localized (Vegfa: 2.32 ± 0.50 at 7 days and 4.37 ± 0.75 at 14 days; Vwf: 4.13 ± 0.82 and 7.74 ± 0.91; Col3a1: 5.43 ± 0.37 and 6.66 ± 0.89). Altogether, our data supported the conclusion that the localized delivery of PDGF-BB from PLGA-MSVs induced the localized de novo formation of fully functional vessels in vivo. With this study, we demonstrated that PLGA-MSV holds promise for accomplishing the controlled localized in vivo release of GFs for the design of innovative tissue engineering strategies.
Collapse
Affiliation(s)
| | - Laura Pandolfi
- College of Materials Science and Engineering, University of Chinese Academy of Science , 19A Yuquanlu, Beijing 100049, China
| | | | | | | | | | | | | | - Ennio Tasciotti
- Department of Orthopedics, Houston Methodist Hospital , 6565 Fannin Street, Houston, Texas 77030, United States
| |
Collapse
|
58
|
Singh A, Trivedi P, Jain NK. Advances in siRNA delivery in cancer therapy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:274-283. [PMID: 28423924 DOI: 10.1080/21691401.2017.1307210] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
RNA interference (RNAi)-based therapeutic approaches are under vibrant scrutinisation to seek cancer cure. siRNA suppress expression of the carcinogenic genes by targeting the mRNA expression. However, in vivo systemic siRNA therapy is hampered by the barriers such as poor cellular uptake, instability under physiological conditions, off-target effects and possible immunogenicity. To overcome these challenges, systemic siRNA therapy warrants the development of clinically suitable, safe, and effective drug delivery systems. Herein, we review the barriers, potential siRNA drug delivery systems, and application of siRNA in clinical trials for cancer therapy. Further research is required to harness the full potential of siRNA as a cancer therapeutic.
Collapse
Affiliation(s)
- Aishwarya Singh
- a School of Pharmaceutical Sciences, Rajiv Gandhi Technical University , Bhopal , Madhya Pradesh , India
| | - Piyush Trivedi
- a School of Pharmaceutical Sciences, Rajiv Gandhi Technical University , Bhopal , Madhya Pradesh , India
| | - Narendra Kumar Jain
- a School of Pharmaceutical Sciences, Rajiv Gandhi Technical University , Bhopal , Madhya Pradesh , India
| |
Collapse
|
59
|
Lee JY, Crake C, Teo B, Carugo D, de Saint Victor M, Seth A, Stride E. Ultrasound-Enhanced siRNA Delivery Using Magnetic Nanoparticle-Loaded Chitosan-Deoxycholic Acid Nanodroplets. Adv Healthc Mater 2017; 6. [PMID: 28195673 DOI: 10.1002/adhm.201601246] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/07/2017] [Indexed: 12/14/2022]
Abstract
Small interfering RNA (siRNA) has significant therapeutic potential but its clinical translation has been severely inhibited by a lack of effective delivery strategies. Previous work has demonstrated that perfluorocarbon nanodroplets loaded with magnetic nanoparticles can facilitate the intracellular delivery of a conventional chemotherapeutic drug. The aim of this study is to determine whether a similar agent can provide a means of delivering siRNA, enabling efficient transfection without degradation of the molecule. Chitosan-deoxycholic acid nanoparticles containing perfluoropentane and iron oxide (d 0 = 7.5 ± 0.35 nm) with a mean hydrodynamic diameter of 257.6 ± 10.9 nm are produced. siRNA (AllStars Hs cell death siRNA) is electrostatically bound to the particle surface and delivery to lung cancer cells and breast cancer cells is investigated with and without ultrasound exposure (500 kHz, 1 MPa peak-to-peak focal pressure, 40 cycles per burst, 1 kHz pulse repetition frequency, 10 s duration). The results show that siRNA functionality is not impaired by the treatment protocol and that the nanodroplets are able to successfully promote siRNA uptake, leading to significant apoptosis (52.4%) 72 h after ultrasound treatment.
Collapse
Affiliation(s)
- Jeong Yu Lee
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| | - Calum Crake
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| | - Boon Teo
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| | - Dario Carugo
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
- Faculty of Engineering and the Environment; Southampton University; Southampton SO17 1BJ UK
| | - Marie de Saint Victor
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| | - Anjali Seth
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| | - Eleanor Stride
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| |
Collapse
|
60
|
Wareing N, Szymanski K, Akkaraju GR, Loni A, Canham LT, Gonzalez-Rodriguez R, Coffer JL. In Vitro Gene Delivery with Large Porous Silicon Nanoparticles Fabricated Using Cost-Effective, Metal-Assisted Chemical Etching. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1602739. [PMID: 28084695 DOI: 10.1002/smll.201602739] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 09/23/2016] [Indexed: 05/28/2023]
Abstract
The cytocompatibility, cell membrane affinity, and plasmid DNA delivery from surface oxidized, metal-assisted stain-etched mesoporous silicon nanoscale particles (pSiNPs) to human embryonic kidney (HEK293) cells is demonstrated, suggesting the possibility of using such material for targeted transfection and drug delivery.
Collapse
Affiliation(s)
- Nancy Wareing
- Department of Biology, Texas Christian University, Fort Worth, TX, 76129, USA
| | - Kyle Szymanski
- Department of Biology, Texas Christian University, Fort Worth, TX, 76129, USA
| | - Giridhar R Akkaraju
- Department of Biology, Texas Christian University, Fort Worth, TX, 76129, USA
| | - Armando Loni
- pSiMedica Ltd, Malvern Hills Science Park, Geraldine Road, Malvern, Worcestershire, WR14 3 SZ, UK
| | - Leigh T Canham
- pSiMedica Ltd, Malvern Hills Science Park, Geraldine Road, Malvern, Worcestershire, WR14 3 SZ, UK
| | | | - Jeffery L Coffer
- Department of Chemistry, Texas Christian University, Fort Worth, TX, 76129, USA
| |
Collapse
|
61
|
Li Y, Wang H, Wang K, Hu Q, Yao Q, Shen Y, Yu G, Tang G. Targeted Co-delivery of PTX and TR3 siRNA by PTP Peptide Modified Dendrimer for the Treatment of Pancreatic Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1602697. [PMID: 27762495 DOI: 10.1002/smll.201602697] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Indexed: 06/06/2023]
Abstract
A new type of tumor-targeted nanovehicle peptide-conjugated PSPG (PSPGP) is successfully synthesized for co-delivery of paclitaxel (PTX) and TR3 small interfering RNA (siRNA). In vitro and in vivo investigations demonstrate that the redox-responsive PSPGP exhibit enhanced endosomal escape and intracellular degradation, which facilitate PTX and TR3 siRNA release, effectively improving the antitumor efficacy.
Collapse
Affiliation(s)
- Yang Li
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Hebin Wang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
- College of Life Sciences, Tarim University, Alar, 843300, China
| | - Kai Wang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Qinglian Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Qi Yao
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Youqing Shen
- Center for Bionanoengineering and State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Guocan Yu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Guping Tang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| |
Collapse
|
62
|
Zhao YN, Piao YZ, Zhang CM, Jiang YM, Liu A, Cui SH, Zhi DF, Zhen YH, Zhang SB. Replacement of quaternary ammonium headgroups by tri-ornithine in cationic lipids for the improvement of gene delivery in vitro and in vivo. J Mater Chem B 2017; 5:7963-7973. [DOI: 10.1039/c7tb01915g] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Replacement of quaternary ammonium headgroups by tri-ornithine in lipids improved gene delivery in vitro and in vivo with little toxicity.
Collapse
Affiliation(s)
- Y. N. Zhao
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education
- Dalian Minzu University
- Dalian
- China
| | - Y. Z. Piao
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education
- Dalian Minzu University
- Dalian
- China
| | - C. M. Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education
- Dalian Minzu University
- Dalian
- China
| | - Y. M. Jiang
- College of Phamacy
- Dalian Medical University
- Dalian
- China
| | - A. Liu
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education
- Dalian Minzu University
- Dalian
- China
| | - S. H. Cui
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education
- Dalian Minzu University
- Dalian
- China
| | - D. F. Zhi
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education
- Dalian Minzu University
- Dalian
- China
| | - Y. H. Zhen
- College of Phamacy
- Dalian Medical University
- Dalian
- China
| | - S. B. Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education
- Dalian Minzu University
- Dalian
- China
| |
Collapse
|
63
|
Deng Q, Li X, Zhu L, He H, Chen D, Chen Y, Yin L. Serum-resistant, reactive oxygen species (ROS)-potentiated gene delivery in cancer cells mediated by fluorinated, diselenide-crosslinked polyplexes. Biomater Sci 2017; 5:1174-1182. [DOI: 10.1039/c7bm00334j] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Fluorinated, diselenide-crosslinked polyplexes were developed to enable ROS-responsive and serum-resistant gene delivery in cancer cells.
Collapse
Affiliation(s)
- Qiurong Deng
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- P.R. China
| | - Xudong Li
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- P.R. China
| | - Lipeng Zhu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- P.R. China
| | - Hua He
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- P.R. China
| | - Donglai Chen
- Department of Thoracic Surgery
- Shanghai Pulmonary Hospital
- Tongji University School of Medicine
- Shanghai
- P.R. China
| | - Yongbing Chen
- Department of Cardiothoracic Surgery
- The Second Affiliated Hospital of Soochow University
- Suzhou 215004
- P.R. China
| | - Lichen Yin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- P.R. China
| |
Collapse
|
64
|
Wong WT, Ma S, Tian XY, Gonzalez AB, Ebong EE, Shen H. Targeted Delivery of Shear Stress-Inducible Micrornas by Nanoparticles to Prevent Vulnerable Atherosclerotic Lesions. Methodist Debakey Cardiovasc J 2016; 12:152-156. [PMID: 27826369 DOI: 10.14797/mdcj-12-3-152] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular wall disease, and endothelial cell dysfunction plays an important role in its development and progression. Under the influence of laminar shear stress, however, the endothelium releases homeostatic factors such as nitric oxide and expresses of vasoprotective microRNAs that are resistant to atherosclerosis. Adhesion molecules such as E-selectin, exhibited on the endothelial surface, recruit monocytes that enter the vessel wall to form foam cells. Accumulation of these foam cells form fatty streaks that may progress to atherosclerotic plaques in the blood vessel wall. Interestingly, E-selectin may also serve as an affinity moiety for targeted drug delivery against atherosclerosis. We have recently developed an E-selectin-targeted platform that enriches therapeutic microRNAs in the inflamed endothelium to inhibit formation of vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Wing Tak Wong
- Houston Methodist Research Institute, Houston, Texas
| | - Shuangtao Ma
- Houston Methodist Research Institute, Houston, Texas
| | - Xiao Yu Tian
- Houston Methodist Research Institute, Houston, Texas
| | | | - Eno E Ebong
- Northeastern University, Boston, Massachusetts
| | - Haifa Shen
- Houston Methodist Research Institute, Houston, Texas
| |
Collapse
|
65
|
RNA interference mediated downregulation of human telomerase reverse transcriptase (hTERT) in LN18 cells. Cytotechnology 2016; 68:2311-2321. [PMID: 27757712 DOI: 10.1007/s10616-016-0025-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 08/24/2016] [Indexed: 10/20/2022] Open
Abstract
Human telomerase reverse transcriptase (hTERT) gene is a biomarker for the targeted therapy in various cancers. Presence of increased telomerase activity is a common feature of all cancers including glioblastoma. Both RNA and catalytic subunits of hTERT are the target sites for blocking its activity. The current study focuses on the expression of hTERT in glioblastoma and its regulation using two different novel siRNAs (small interfering RNA). Our patient data demonstrated increased expression of hTERT, which could be correlated with carcinogenesis in glioma. In vitro studies in siRNA transfected LN18 cells confirmed significant cell death (p < 0.05) as evidenced by MTT and trypan blue exclusion assay. These results were further supported by flow cytometry data, which showed significant increase in early and late apoptosis. The hTERT mRNA expression was effectively downregulated by 45 and 39 % with siRNA1 and siRNA2, respectively. These results were further confirmed by immunoblotting analysis (p < 0.05). Our results suggest that both the siRNAs effectively down regulated the expression of hTERT at mRNA and protein levels, thereby decreasing cell viability and proliferation rate. Hence siRNA mediated downregulation of hTERT could be a potential therapeutic avenue in glioblastoma.
Collapse
|
66
|
Li Y, Huang X, Lee RJ, Qi Y, Wang K, Hao F, Zhang Y, Lu J, Meng Q, Li S, Xie J, Teng L. Synthesis of Polymer-Lipid Nanoparticles by Microfluidic Focusing for siRNA Delivery. Molecules 2016; 21:E1314. [PMID: 27763492 PMCID: PMC6274485 DOI: 10.3390/molecules21101314] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 08/13/2016] [Accepted: 09/20/2016] [Indexed: 01/09/2023] Open
Abstract
Polyethylenimine (PEI) as a cationic polymer is commonly used as a carrier for gene delivery. PEI-800 is less toxic than PEI-25K but it is also less efficient. A novel nanocarrier was developed by combining PEI-800 with a pH-sensitive lipid to form polymer-lipid hybrid nanoparticles (P/LNPs). They were synthesized by microfluidic focusing (MF). Two microfluidic devices were used to synthesize P/LNPs loaded with VEGF siRNA. A series of P/LNPs with different particle sizes and distributions were obtained by altering the flow rate and geometry of microfluidic chips, and introducing sonication. Furthermore, the P/LNPs can be loaded with VEGF siRNA efficiently and were stable in serum for 12 h. Finally, P/LNPs produced by the microfluidic chip showed greater cellular uptake as well as down-regulation of VEGF protein level in both A549 and MCF-7 with reduced cellular toxicity. All in all, the P/LNPs produced by MF method were shown to be a safe and efficient carrier for VEGF siRNA, with potential application for siRNA therapeutics.
Collapse
Affiliation(s)
- Yujing Li
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
| | - Xueqin Huang
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
- Department of Chemistry and Pharmacy, Zhuhai College of Jilin University, Zhuhai 519041, China.
| | - Robert J Lee
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.
| | - Yuhang Qi
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
| | - Kaikai Wang
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
| | - Fei Hao
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
| | - Yu Zhang
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
| | - Jiahui Lu
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
| | - Qingfan Meng
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
| | - Shuai Li
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
| | - Jing Xie
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Qianjin Street No. 2699, Changchun 130012, China.
| |
Collapse
|
67
|
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
|
68
|
Scott B, Shen J, Nizzero S, Boom K, Persano S, Mi Y, Liu X, Zhao Y, Blanco E, Shen H, Ferrari M, Wolfram J. A pyruvate decarboxylase-mediated therapeutic strategy for mimicking yeast metabolism in cancer cells. Pharmacol Res 2016; 111:413-421. [PMID: 27394167 DOI: 10.1016/j.phrs.2016.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/28/2016] [Accepted: 07/05/2016] [Indexed: 01/18/2023]
Abstract
Cancer cells have high rates of glycolysis and lactic acid fermentation in order to fuel accelerated rates of cell division (Warburg effect). Here, we present a strategy for merging cancer and yeast metabolism to remove pyruvate, a key intermediate of cancer cell metabolism, and produce the toxic compound acetaldehyde. This approach was achieved by administering the yeast enzyme pyruvate decarboxylase to triple negative breast cancer cells. To overcome the challenges of protein delivery, a nanoparticle-based system consisting of cationic lipids and porous silicon were employed to obtain efficient intracellular uptake. The results demonstrate that the enzyme therapy decreases cancer cell viability through production of acetaldehyde and reduction of lactic acid fermentation.
Collapse
Affiliation(s)
- Bronwyn Scott
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jianliang Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Sara Nizzero
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Applied Physics Graduate Program, Rice University, Houston, TX 77005, USA
| | - Kathryn Boom
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Stefano Persano
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Yu Mi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China; Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
69
|
Movassaghian S, Xie Y, Hildebrandt C, Rosati R, Li Y, Kim NH, Conti DS, da Rocha SRP, Yang ZQ, Merkel OM. Post-Transcriptional Regulation of the GASC1 Oncogene with Active Tumor-Targeted siRNA-Nanoparticles. Mol Pharm 2016; 13:2605-21. [PMID: 27223606 DOI: 10.1021/acs.molpharmaceut.5b00948] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Basal-like breast cancer (BLBC) accounts for the most aggressive types of breast cancer, marked by high rates of relapse and poor prognoses and with no effective clinical therapy yet. Therefore, investigation of new targets and treatment strategies is more than necessary. Here, we identified a receptor that can be targeted in BLBC for efficient and specific siRNA mediated gene knockdown of therapeutically relevant genes such as the histone demethylase GASC1, which is involved in multiple signaling pathways leading to tumorigenesis. Breast cancer and healthy breast cell lines were compared regarding transferrin receptor (TfR) expression via flow cytometry and transferrin binding assays. Nanobioconjugates made of low molecular weight polyethylenimine (LMW-PEI) and transferrin (Tf) were synthesized to contain a bioreducible disulfide bond. siRNA complexation was characterized by condensation assays and dynamic light scattering. Cytotoxicity, transfection efficiency, and the targeting specificity of the conjugates were investigated in TfR positive and negative healthy breast and breast cancer cell lines by flow cytometry, confocal microscopy, RT-PCR, and Western blot. Breast cancer cell lines revealed a significantly higher TfR expression than healthy breast cells. The conjugates efficiently condensed siRNA into particles with 45 nm size at low polymer concentrations, showed no apparent toxicity on different breast cancer cell lines, and had significantly greater transfection and gene knockdown activity on mRNA and protein levels than PEI/siRNA leading to targeted and therapeutic growth inhibition post GASC1 knockdown. The synthesized nanobioconjugates improved the efficiency of gene transfer and targeting specificity in transferrin receptor positive cells but not in cells with basal receptor expression. Therefore, these materials in combination with our newly identified siRNA sequences are promising candidates for therapeutic targeting of hard-to-treat BLBC and are currently further investigated regarding in vivo targeting efficacy and biocompatibility.
Collapse
Affiliation(s)
- Sara Movassaghian
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States
| | - Yuran Xie
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States
| | - Claudia Hildebrandt
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Pharmaceutics and Biopharmaceutics, Kiel University , 24118 Kiel, Germany
| | - Rayna Rosati
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States
| | - Ying Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States
| | - Na Hyung Kim
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States
| | - Denise S Conti
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University , Detroit, Michigan 48202, United States
| | - Sandro R P da Rocha
- Department of Pharmaceutics, College of Pharmacy, Virginia Commonwealth University , Richmond, Virginia 23298, United States.,Department of Chemical and Life Science Engineering, Virginia Commonwealth University , Richmond, Virginia 23284, United States
| | - Zeng-Quan Yang
- Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States
| | - Olivia M Merkel
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States.,Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München , 80539 Munich, Germany
| |
Collapse
|
70
|
Modulated cellular delivery of anti-VEGF siRNA (bevasiranib) by incorporating supramolecular assemblies of hydrophobically modified polyamidoamine dendrimer in stealth liposomes. Int J Pharm 2016; 510:30-41. [PMID: 27291973 DOI: 10.1016/j.ijpharm.2016.06.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 12/16/2022]
Abstract
A novel lipopolymer based system was designed and characterized for cellular delivery of anti-VEGF siRNA in SKBR-3 breast tumor cell line. Polyamidoamine (PAMAM) dendrimers of low generations (G1, G2 and G3) were incorporated into polyethylene glycol (PEG)-stabilized liposomes by following the consecutive steps: (a) synthesis of the cholesterol conjugates (40% molar ratio of cholesterol to primary amines of PAMAM), (b) incorporation of the conjugates in liposome by lipid mixing and (c) microencapsulation of the siRNA using the ethanol drop method. The cholesterol conjugates of PAMAM dendrimers (G1-Chol40%, G2-Chol40% and G3-Chol40%) formed self assembly with low CMC values (<11μg/ml). Not only did G2-Chol40% show the highest lipid mixing among the cholesterol conjugates, but also, had the lowest leakage of encapsulated carboxyfluorescein tracer. Various N(amine))/L(lipid)/P(phosphate) mole ratios were investigated for siRNA condensation by ethidium bromide dye exclusion assay. The optimum N/L/P ratio of 20:33:10 was chosen for microencapsulation of anti-VEGF siRNA by ethanol drop method, showing particle size of 130nm, zeta-potential of +4mV, siRNA loading efficiency and capacity of 96% and 13wt%, and high stability against heparin sulfate (extracellular matrix). TEM shows uniform and discrete oligo- or multi-lamellar vesicular structures. The liposome incorporating G2-Chol40% was successfully internalized into SKBR-3 cells mainly through clathrin-mediated endocytosis, which was able to escape from endosomes and showed a significantly higher sequence-specific inhibition of VEGF expression and cell growth than the respective G2-Chol40%/siRNA dendriplexes. Importantly, the cytotoxicity decreased with incorporation of G2-Chol40% in the liposomes.
Collapse
|
71
|
Sharma N, Jha S. NLR-regulated pathways in cancer: opportunities and obstacles for therapeutic interventions. Cell Mol Life Sci 2016; 73:1741-64. [PMID: 26708292 PMCID: PMC11108278 DOI: 10.1007/s00018-015-2123-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/09/2015] [Accepted: 12/15/2015] [Indexed: 02/08/2023]
Abstract
NLRs (nucleotide-binding domain, leucine-rich repeat containing receptors) are pattern recognition receptors associated with immunity and inflammation in response to endogenous and exogenous pathogen and damage associated molecular patterns (PAMPs and DAMPs respectively). Dysregulated NLR function is associated with several diseases including cancers, metabolic diseases, autoimmune disorders and autoinflammatory syndromes. In the last decade, distinct cell and organ specific roles for NLRs have been identified however; their roles in cancer initiation, development and progression remain controversial. This review summarizes the emerging role of NLRs in cancer and their possible future as targets for cancer therapeutics.
Collapse
Affiliation(s)
- Nidhi Sharma
- Department of Biology, Indian Institute of Technology Jodhpur, Old Residency Road, Ratanada, Jodhpur, Rajasthan, 342011, India
| | - Sushmita Jha
- Department of Biology, Indian Institute of Technology Jodhpur, Old Residency Road, Ratanada, Jodhpur, Rajasthan, 342011, India.
| |
Collapse
|
72
|
|
73
|
Friberg S, Nyström AM. NANOMEDICINE: will it offer possibilities to overcome multiple drug resistance in cancer? J Nanobiotechnology 2016; 14:17. [PMID: 26955956 PMCID: PMC4784447 DOI: 10.1186/s12951-016-0172-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 03/03/2016] [Indexed: 12/12/2022] Open
Abstract
This review is written with the purpose to review the current nanomedicine literature and provide an outlook on the developments in utilizing nanoscale drug constructs in treatment of solid cancers as well as in the potential treatment of multi-drug resistant cancers. No specific design principles for this review have been utilized apart from our active choice to avoid results only based on in vitro studies. Few drugs based on nanotechnology have progressed to clinical trials, since most are based only on in vitro experiments which do not give the necessary data for the research to progress towards pre-clinical studies. The area of nanomedicine has indeed spark much attention and holds promise for improved future therapeutics in the treatment of solid cancers. However, despite much investment few targeted therapeutics have successfully progressed to early clinical trials, indicating yet again that the human body is complicated and that much more understanding of the fundamentals of receptor interactions, physics of nanomedical constructs and their circulation in the body is indeed needed. We believe that nanomedical therapeutics can allow for more efficient treatments of resistant cancers, and may well be a cornerstone for RNA based therapeutics in the future given their general need for shielding from the harsh environment in the blood stream.
Collapse
Affiliation(s)
- Sten Friberg
- Department of Neuroscience, Swedish Medical Nanoscience Center, Karolinska Institutet, Retzius väg 8, 171 77, Stockholm, Sweden.
| | - Andreas M Nyström
- Institute of Environmental Medicine, Karolinska Institutet, Nobels väg 13, 171 77, Stockholm, Sweden.
| |
Collapse
|
74
|
Chen Z, Krishnamachary B, Bhujwalla ZM. Degradable Dextran Nanopolymer as a Carrier for Choline Kinase (ChoK) siRNA Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2016; 6:E34. [PMID: 28344291 PMCID: PMC5302479 DOI: 10.3390/nano6020034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/19/2016] [Accepted: 02/04/2016] [Indexed: 01/28/2023]
Abstract
Although small interfering RNA (siRNA) therapy has proven to be a specific and effective treatment in cells, the delivery of siRNA is a challenge for the applications of siRNA therapy. We present a degradable dextran with amine groups as an siRNA nano-carrier. In our nano-carrier, the amine groups are conjugated to the dextran platform through the acetal bonds, which are acid sensitive. Therefore this siRNA carrier is stable in neutral and basic conditions, while the amine groups can be cleaved and released from dextran platform under weak acid conditions (such as in endosomes). The cleavage and release of amine groups can reduce the toxicity of cationic polymer and enhance the transfection efficiency. We successfully applied this nano-carrier to deliver choline kinase (ChoK) siRNA for ChoK inhibition in cells.
Collapse
Affiliation(s)
- Zhihang Chen
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Balaji Krishnamachary
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Zaver M Bhujwalla
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
75
|
Molla MR, Levkin PA. Combinatorial Approach to Nanoarchitectonics for Nonviral Delivery of Nucleic Acids. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:1159-1175. [PMID: 26608939 DOI: 10.1002/adma.201502888] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/01/2015] [Indexed: 06/05/2023]
Abstract
Nanoparticles based on cationic polymers, lipids or lipidoids are of great interest in the field of gene delivery applications. The research on these nanosystems is rapidly growing as they hold promise to treat wide variety of human diseases ranging from viral infections to genetic disorders and cancer. Recently, combinatorial design principles have been adopted for rapid generation of large numbers of chemically diverse polymers and lipids capable of forming multifunctional nanocarriers for the use in gene delivery applications. At the same time, current high-throughput screening systems as well as convenient cell assays and readout techniques allow for fast evaluation of cell transfection efficiencies and toxicities of libraries of novel gene delivery agents. This allows for a rapid evaluation of structure-function relationship as well as identification of novel efficient nanocarriers for cell transfection and gene therapy. Here, the recent contribution of high-throughput synthesis to the development of novel nanocarriers for gene delivery applications is described.
Collapse
Affiliation(s)
- Mijanur Rahaman Molla
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Pavel A Levkin
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
- University of Heidelberg, Department of Applied Physical Chemistry, 69120, Heidelberg, Germany
| |
Collapse
|
76
|
Alagia A, Eritja R. siRNA and RNAi optimization. WILEY INTERDISCIPLINARY REVIEWS-RNA 2016; 7:316-29. [PMID: 26840434 DOI: 10.1002/wrna.1337] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 12/12/2022]
Abstract
The discovery and examination of the posttranscriptional gene regulatory mechanism known as RNA interference (RNAi) contributed to the identification of small interfering RNA (siRNA) and the comprehension of its enormous potential for clinical purposes. Theoretically, the ability of specific target gene downregulation makes the RNAi pathway an appealing solution for several diseases. Despite numerous hurdles resulting from the inherent properties of siRNA molecule and proper delivery to the target tissue, more than 50 RNA-based drugs are currently under clinical testing. In this work, we analyze the recent literature in the optimization of siRNA molecules. In detail, we focused on describing the most recent advances of siRNA field aimed at optimize siRNA pharmacokinetic properties. Special attention has been given in describing the impact of RNA modifications in the potential off-target effects (OTEs) such as saturation of the RNAi machinery, passenger strand-mediated silencing, immunostimulation, and miRNA-like OTEs as well as to recent developments on the delivery issue. The novel delivery systems and modified siRNA provide significant steps toward the development of reliable siRNA molecules for therapeutic use. WIREs RNA 2016, 7:316-329. doi: 10.1002/wrna.1337 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Adele Alagia
- Chemical and Biomolecular Nanotechnology, CIBER-BBN, Institute for Advanced Chemistry of Catalonia, IQAC-CSIC, Barcelona, Spain
| | - Ramon Eritja
- Chemical and Biomolecular Nanotechnology, CIBER-BBN, Institute for Advanced Chemistry of Catalonia, IQAC-CSIC, Barcelona, Spain
| |
Collapse
|
77
|
Sato T, Sato Y, Iwai K, Kuge S, Teramae N, Nishizawa S. Fluorescence imaging of siRNA delivery by peptide nucleic acid-based probe. ANAL SCI 2016; 31:315-20. [PMID: 25864675 DOI: 10.2116/analsci.31.315] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We report on the use of a peptide nucleic acid (PNA)-based fluorescent probe for the analysis of siRNA delivery to living cells. The probe, Py-AA-TO, possesses thiazole orange (TO) and pyrene moieties in the C- and N-termini of PNA, and can function as a light-up probe capable of selective binding to 3'-overhanging nucleotides of target siRNAs. The affinity-labeling of the siRNAs with Py-AA-TO facilitates fluorescence imaging of cellular uptake of polymer-based carriers encapsulating the siRNAs (polyplexes) through endocytosis and subsequent sequestration into lysosome. In addition, flow cytometric measurements reveal that the monitoring of Py-AA-TO fluorescence inside the cells is successfully applicable to the analysis of the polyplex disassembly. These promising functions of Py-AA-TO are presented and discussed as a basis for the design of molecular probes for fluorescent imaging and quantitative analysis of the siRNA delivery process.
Collapse
Affiliation(s)
- Takaya Sato
- Department of Chemistry, Graduate School of Science, Tohoku University
| | | | | | | | | | | |
Collapse
|
78
|
Liu Y, Wang J. Therapeutic Potentials of Noncoding RNAs: Targeted Delivery of ncRNAs in Cancer Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 927:429-58. [PMID: 27376745 DOI: 10.1007/978-981-10-1498-7_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Knowledge of multiple actions of short noncoding RNAs (ncRNAs) has truly allowed for viewing DNA, RNA, and protein in novel ways. The ncRNAs are an attractive new class of therapeutics, especially against undruggable targets for the treatment of cancer and other diseases. Despite the potential of ncRNAs in cancer therapy, many challenges remain, including rapid degradation and clearance, poor cellular uptake, off-target effects, and immunogenicity. Rational design, chemical modifications, and delivery carriers offer significant opportunities to overcome these challenges. In this chapter, the development of ncRNAs as cancer therapeutics from early stages to clinical trials and strategies for ncRNA-targeted delivery to cancer cells will be introduced.
Collapse
Affiliation(s)
- Yang Liu
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, People's Republic of China
| | - Jun Wang
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, People's Republic of China.
| |
Collapse
|
79
|
Pandolfi L, Minardi S, Taraballi F, Liu X, Ferrari M, Tasciotti E. Composite microsphere-functionalized scaffold for the controlled release of small molecules in tissue engineering. J Tissue Eng 2016; 7:2041731415624668. [PMID: 26977286 PMCID: PMC4765809 DOI: 10.1177/2041731415624668] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/20/2015] [Indexed: 12/17/2022] Open
Abstract
Current tissue engineering strategies focus on restoring damaged tissue architectures using biologically active scaffolds. The ideal scaffold would mimic the extracellular matrix of any tissue of interest, promoting cell proliferation and de novo extracellular matrix deposition. A plethora of techniques have been evaluated to engineer scaffolds for the controlled and targeted release of bioactive molecules to provide a functional structure for tissue growth and remodeling, as well as enhance recruitment and proliferation of autologous cells within the implant. Recently, novel approaches using small molecules, instead of growth factors, have been exploited to regulate tissue regeneration. The use of small synthetic molecules could be very advantageous because of their stability, tunability, and low cost. Herein, we propose a chitosan-gelatin scaffold functionalized with composite microspheres consisting of mesoporous silicon microparticles and poly(dl-lactic-co-glycolic acid) for the controlled release of sphingosine-1-phospate, a small molecule of interest. We characterized the platform with scanning electron microscopy, Fourier transform infrared spectroscopy, and confocal microscopy. Finally, the biocompatibility of this multiscale system was analyzed by culturing human mesenchymal stem cells onto the scaffold. The presented strategy establishes the basis of a versatile scaffold for the controlled release of small molecules and for culturing mesenchymal stem cells for regenerative medicine applications.
Collapse
Affiliation(s)
- Laura Pandolfi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
- College of Materials Science and Engineering, University of Chinese Academy of Science, Beijing, China
| | - Silvia Minardi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Francesca Taraballi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Xeuwu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Ennio Tasciotti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| |
Collapse
|
80
|
Wolfram J, Shen H, Ferrari M. Multistage vector (MSV) therapeutics. J Control Release 2015; 219:406-415. [PMID: 26264836 PMCID: PMC4656100 DOI: 10.1016/j.jconrel.2015.08.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/01/2015] [Accepted: 08/04/2015] [Indexed: 12/21/2022]
Abstract
One of the greatest challenges in the field of medicine is obtaining controlled distribution of systemically administered therapeutic agents within the body. Indeed, biological barriers such as physical compartmentalization, pressure gradients, and excretion pathways adversely affect localized delivery of drugs to pathological tissue. The diverse nature of these barriers requires the use of multifunctional drug delivery vehicles that can overcome a wide range of sequential obstacles. In this review, we explore the role of multifunctionality in nanomedicine by primarily focusing on multistage vectors (MSVs). The MSV is an example of a promising therapeutic platform that incorporates several components, including a microparticle, nanoparticles, and small molecules. In particular, these components are activated in a sequential manner in order to successively address transport barriers.
Collapse
Affiliation(s)
- Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
81
|
Yu X, Yang G, Shi Y, Su C, Liu M, Feng B, Zhao L. Intracellular targeted co-delivery of shMDR1 and gefitinib with chitosan nanoparticles for overcoming multidrug resistance. Int J Nanomedicine 2015; 10:7045-56. [PMID: 26648717 PMCID: PMC4648604 DOI: 10.2147/ijn.s92436] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Nowadays, multidrug resistance and side effects of drugs limit the effectiveness of chemotherapies in clinics. P-glycoprotein (P-gp) (MDR1), as a member of the ATP-binding cassette family, acts on transporting drugs into cell plasma across the membrane of cancer cells and leads to the occurrence of multidrug resistance, thus resulting in the failure of chemotherapy in cancer. The main aims of this research were to design a nanodelivery system for accomplishing the effective co-delivery of gene and antitumor drug and overcoming multidrug resistance effect. In this study, shMDR1 and gefitinib-encapsulating chitosan nanoparticles with sustained release, small particle size, and high encapsulation efficiency were prepared. The serum stability, protection from nuclease, and transfection efficiency of gene in vitro were investigated. The effects of co-delivery of shMDR1 and gefitinib in nanoparticles on reversing multidrug resistance were also evaluated by investigating the cytotoxicity, cellular uptake mechanism, and cell apoptosis on established gefitinib-resistant cells. The results demonstrated that chitosan nanoparticles entrapping gefitinib and shMDR1 had the potential to overcome the multidrug resistance and improve cancer treatment efficacy, especially toward resistant cells.
Collapse
Affiliation(s)
- Xiwei Yu
- School of Pharmacy, Liaoning Medical University, Jinzhou, People’s Republic of China
| | - Guang Yang
- Department of Oncology, BenQ Medical Center, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yijie Shi
- School of Pharmacy, Liaoning Medical University, Jinzhou, People’s Republic of China
| | - Chang Su
- School of Veterinary Medicine, Liaoning Medical University, Jinzhou, People’s Republic of China
| | - Ming Liu
- School of Pharmacy, Liaoning Medical University, Jinzhou, People’s Republic of China
| | - Bo Feng
- School of Pharmacy, Liaoning Medical University, Jinzhou, People’s Republic of China
| | - Liang Zhao
- School of Pharmacy, Liaoning Medical University, Jinzhou, People’s Republic of China
| |
Collapse
|
82
|
Tekie FSM, Atyabi F, Soleimani M, Arefian E, Atashi A, Kiani M, Khoshayand MR, Amini M, Dinarvand R. Chitosan polyplex nanoparticle vector for miR-145 expression in MCF-7: Optimization by design of experiment. Int J Biol Macromol 2015; 81:828-37. [DOI: 10.1016/j.ijbiomac.2015.09.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/11/2015] [Accepted: 09/08/2015] [Indexed: 01/13/2023]
|
83
|
Freire JEDC, Medeiros SCD, Lopes Neto AV, Monteiro Júnior JE, Sousa AJS, Rocha AJ, Menezes LMBD. Bioethical conflicts of gene therapy: a brief critical review. Rev Assoc Med Bras (1992) 2015; 60:520-4. [PMID: 25650850 DOI: 10.1590/1806-9282.60.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 06/03/2014] [Indexed: 11/22/2022] Open
Abstract
Methods and techniques employed in gene therapy are reviewed in parallel with pertinent ethical conflicts. Clinical interventions based on gene therapy techniques preferentially use vectors for the transportation of therapeutic genes, however little is known about the potential risks and damages to the patient. Thus, attending carefully to the clinical complications arising as well as to security is essential. Despite the scientific and technological advances, there are still many uncertainties about the side effects of gene therapy. Moreover, there is a need, above all, to understand the principles of bioethics as both science and ethics, in accordance with its socioecological responsibility, in order to prioritize the health and welfare of man and nature, using properly natural resources and technology. Therefore, it is hard to determine objective results and to which extent the insertion of genes can affect the organism, as well as the ethical implication.
Collapse
Affiliation(s)
| | | | - Antônio Viana Lopes Neto
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | | | - Antônio José Rocha
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, CE, Brazil
| | | |
Collapse
|
84
|
Pan R, Xu W, Yuan F, Chu D, Ding Y, Chen B, Jafari M, Yuan Y, Chen P. A novel peptide for efficient siRNA delivery in vitro and therapeutics in vivo. Acta Biomater 2015; 21:74-84. [PMID: 25861950 DOI: 10.1016/j.actbio.2015.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 03/18/2015] [Accepted: 04/02/2015] [Indexed: 12/31/2022]
Abstract
Small interfering RNA (siRNA) shows great therapeutic potential due to its ability to regulate gene expression in a highly selective manner. However, its application has been limited by ineffective cellular uptake of siRNAs. To achieve successful gene-silencing efficiency, a safe and effective delivery vector is generally required. In this study, we designed a series of amphipathic peptides that comprised a variant of a nuclear localization sequence, 0-6 histidine residues and an optional stearic acid group. Among these candidates, STR-HK exhibited good characteristics as a safe and efficient siRNA delivery vector, facilitating efficient siRNA delivery to mammalian cells without causing cytotoxicity. Moreover, the intratumoral injection of STR-HK/siRNA complexes achieved high anti-tumor activity through the downregulation of the Bcl-2 protein in mice, with an inhibition rate of 62.8%. Our data demonstrate that STR-HK is a highly promising siRNA delivery vector for therapeutic applications.
Collapse
Affiliation(s)
- Ran Pan
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; Waterloo Institute for Nanotechnology, University of Waterloo, ON N2L 3G1, Canada
| | - Wen Xu
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; Waterloo Institute for Nanotechnology, University of Waterloo, ON N2L 3G1, Canada
| | - Feng Yuan
- Department of Pharmacy, Shanghai 3rd People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201999, China
| | - Dafeng Chu
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; Waterloo Institute for Nanotechnology, University of Waterloo, ON N2L 3G1, Canada
| | - Yong Ding
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; Waterloo Institute for Nanotechnology, University of Waterloo, ON N2L 3G1, Canada
| | - Baoling Chen
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; Waterloo Institute for Nanotechnology, University of Waterloo, ON N2L 3G1, Canada
| | - Mousa Jafari
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; Waterloo Institute for Nanotechnology, University of Waterloo, ON N2L 3G1, Canada
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai 3rd People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201999, China
| | - P Chen
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; Waterloo Institute for Nanotechnology, University of Waterloo, ON N2L 3G1, Canada.
| |
Collapse
|
85
|
Friberg S, Nyström AM. Nanotechnology in the war against cancer: new arms against an old enemy – a clinical view. Future Oncol 2015; 11:1961-75. [DOI: 10.2217/fon.15.91] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
ABSTRACT Clinical oncology is facing a paradigm shift. A new treatment philosophy is emerging and new targets are appearing that require new active agents. The medical use of nanotechnology – nanomedicine – holds several promising possibilities in the war against cancer. Some of these include: new formats for old drugs, that is, increasing efficacy while diminishing side effects; and new administration routes – that is, dermal, oral and pulmonary. In this overview, we describe some nanoparticles and their medical uses as well as highlight advantages of nanoparticles compared with conventional pharmaceuticals. We also point to some of the many technical challenges and potential risks with using nanotechnology for oncological applications.
Collapse
Affiliation(s)
- Sten Friberg
- Swedish Medical Nanoscience Center, Department of Neuroscience, Retzius väg 8, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Andreas M Nyström
- Institute of Environmental Medicine, Nobels väg 13, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
86
|
Parodi A, Corbo C, Cevenini A, Molinaro R, Palomba R, Pandolfi L, Agostini M, Salvatore F, Tasciotti E. Enabling cytoplasmic delivery and organelle targeting by surface modification of nanocarriers. Nanomedicine (Lond) 2015; 10:1923-40. [PMID: 26139126 PMCID: PMC5561781 DOI: 10.2217/nnm.15.39] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nanocarriers are designed to specifically accumulate in diseased tissues. In this context, targeting of intracellular compartments was shown to enhance the efficacy of many drugs and to offer new and more effective therapeutic approaches. This is especially true for therapies based on biologicals that must be encapsulated to favor cell internalization, and to avoid intracellular endosomal sequestration and degradation of the payload. In this review, we discuss specific surface modifications designed to achieve cell cytoplasm delivery and to improve targeting of major organelles; we also discuss the therapeutic applications of these approaches. Last, we describe some integrated strategies designed to sequentially overcome the biological barriers that separate the site of administration from the cell cytoplasm, which is the drug's site of action.
Collapse
Affiliation(s)
- Alessandro Parodi
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
- Fondazione IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Claudia Corbo
- Fondazione IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Armando Cevenini
- Department of Molecular Medicine & Medical Biotechnology, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
- CEINGE, Biotecnologie Avanzate s.c.a.r.l., Via G. Salvatore 486, 80145 Naples, Italy
| | - Roberto Molinaro
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
- Clinica Chirurgica I, Dipartimento di Scienze Chirurgiche Oncologiche e Gastroeterologiche, Università di Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Roberto Palomba
- Fondazione IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Laura Pandolfi
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
- College of Materials Science & Optoelectronic Technology, University of Chinese Academy of Science, 19A Yuquanlu, Beijing, China
| | - Marco Agostini
- Clinica Chirurgica I, Dipartimento di Scienze Chirurgiche Oncologiche e Gastroeterologiche, Università di Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Francesco Salvatore
- Fondazione IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
- CEINGE, Biotecnologie Avanzate s.c.a.r.l., Via G. Salvatore 486, 80145 Naples, Italy
| | - Ennio Tasciotti
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| |
Collapse
|
87
|
Zhang L, Lyer AK, Yang X, Kobayashi E, Guo Y, Mankin H, Hornicek FJ, Amiji MM, Duan Z. Polymeric nanoparticle-based delivery of microRNA-199a-3p inhibits proliferation and growth of osteosarcoma cells. Int J Nanomedicine 2015; 10:2913-24. [PMID: 25931818 PMCID: PMC4404938 DOI: 10.2147/ijn.s79143] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Our prior screening of microRNAs (miRs) identified that miR-199a-3p expression is reduced in osteosarcoma cells, one of the most common types of bone tumor. miR-199a-3p exhibited functions of tumor cell growth inhibition, suggesting the potential application of miR-199a-3p as an anticancer agent. In the study reported here, we designed and developed a lipid-modified dextran-based polymeric nanoparticle platform for encapsulation of miRs, and determined the efficiency and efficacy of delivering miR-199a-3p into osteosarcoma cells. In addition, another potent miR, let-7a, which also displayed tumor suppressive ability, was selected as a candidate miR for evaluation. Fluorescence microscopy studies and real-time polymerase chain reaction results showed that dextran nanoparticles could deliver both miR-199a-3p and let-7a into osteosarcoma cell lines (KHOS and U-2OS) successfully. Western blotting analysis and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays demonstrated that dextran nanoparticles loaded with miRs could efficiently downregulate the expression of target proteins and effectively inhibit the growth and proliferation of osteosarcoma cells. These results demonstrate that a lipid-modified dextran-based polymeric nanoparticle platform may be an effective nonviral carrier for potential miR-based anticancer therapeutics.
Collapse
Affiliation(s)
- Linlin Zhang
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA ; Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Arun K Lyer
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, Massachusetts, USA ; Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Xiaoqian Yang
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eisuke Kobayashi
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Yuqi Guo
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA ; Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Henry Mankin
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, Massachusetts, USA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
88
|
Marson D, Laurini E, Posocco P, Fermeglia M, Pricl S. Cationic carbosilane dendrimers and oligonucleotide binding: an energetic affair. NANOSCALE 2015; 7:3876-3887. [PMID: 25340619 DOI: 10.1039/c4nr04510f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Generation 2 cationic carbosilane dendrimers hold great promise as internalizing agents for gene therapy as they present low toxicity and retain and internalize the genetic material as an oligonucleotide or siRNA. In this work we carried out complete in silico structural and energetical characterization of the interactions of a set of G2 carbosilane dendrimers, showing different affinity towards two single strand oligonucleotide (ODN) sequences in vitro. Our simulations predict that these four dendrimers and the relevant ODN complexes are characterized by similar size and shape, and that the molecule-specific ODN binding ability can be rationalized only by considering a critical molecular design parameter: the normalized effective binding energy ΔG(bind,eff)/N(eff), i.e. the performance of each active individual dendrimer branch directly involved in a binding interaction.
Collapse
Affiliation(s)
- D Marson
- Molecular Simulation Engineering (MOSE) Laboratory, DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy.
| | | | | | | | | |
Collapse
|
89
|
Liu X, Liu C, Zhou J, Chen C, Qu F, Rossi JJ, Rocchi P, Peng L. Promoting siRNA delivery via enhanced cellular uptake using an arginine-decorated amphiphilic dendrimer. NANOSCALE 2015; 7:3867-3875. [PMID: 25283447 DOI: 10.1039/c4nr04759a] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
RNA interference (RNAi) with small interfering RNA (siRNA) is expected to offer an attractive means to specifically and efficiently silence disease-associated genes for treating various diseases provided that safe and efficient delivery systems are available. In this study, we have established an arginine-decorated amphiphilic dendrimer composed of a hydrophobic alkyl chain and a hydrophilic PAMAM dendron bearing arginine terminals as nonviral vector for siRNA delivery. Indeed, this dendrimer proved to be very effective at delivering siRNAs in human prostate cancer PC-3 cells and in human hematopoietic CD34+ stem cells, leading to improved gene silencing compared to the corresponding nonarginine decorated dendrimer. Further investigation confirmed that this dendrimer was granted with the capacity to form stable nanoparticles with siRNA and significantly enhance cellular uptake of siRNA. In addition, this dendrimer revealed no discernible cytotoxicity. All these findings demonstrate that decoration of the dendrimer surface with arginine residues is indeed a useful strategy to improve the delivery ability of dendrimers.
Collapse
Affiliation(s)
- Xiaoxuan Liu
- Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, France.
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Alajangi HK, Santhiya D. Fluorescence and Förster resonance energy transfer investigations on DNA oligonucleotide and PAMAM dendrimer packing interactions in dendriplexes. Phys Chem Chem Phys 2015; 17:8680-91. [PMID: 25738189 DOI: 10.1039/c4cp05295a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Considering the importance of short oligonucleotide packing in dendriplex-mediated gene delivery, a direct insight into the 14-mer oligonucleotide and dendrimer interactions using fluorescence and FRET techniques is the focus of this study. Fluorometric titrations of various fluorophore-tagged oligonucleotides with the first three PAMAM dendrimer generations showed a decrease in the fluorescence intensity with two break points, namely Z and Z, for each titration. The first break point for each dendrimer was identical to the neutralization point observed by basic biophysical studies for the corresponding dendrimer generations. Additionally, FRET studies on dual tagged oligonucleotide (DFT) molecules revealed a third break point at the charge ratio (Z) where there was the highest fluorescence energy transfer from the donor to the acceptor fluorophores. Altogether, dendriplex formation was considered to take place via three steps with an increase in the dendrimer concentration, where initially there was monomeric complexation at the neutralization point (Z) followed by loosely held molecular aggregation of the dendrimer (Z). In the final step, dendrimer molecular aggregates were held tightly together for the closest possible packing of the oligonucleotide molecules onto their surface. The effective molecular packing is identified by the highest FRET intensity for the dendrimer of generation 2 at a charge ratio of 0.34 (Z±).
Collapse
Affiliation(s)
- Hema Kumari Alajangi
- Department of Applied Chemistry and Polymer Technology, Delhi Technological University, Bawana Road, Delhi-110 042, India.
| | | |
Collapse
|
91
|
|
92
|
Zhao X, Li F, Li Y, Wang H, Ren H, Chen J, Nie G, Hao J. Co-delivery of HIF1α siRNA and gemcitabine via biocompatible lipid-polymer hybrid nanoparticles for effective treatment of pancreatic cancer. Biomaterials 2015; 46:13-25. [PMID: 25678112 DOI: 10.1016/j.biomaterials.2014.12.028] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/28/2014] [Accepted: 12/20/2014] [Indexed: 12/17/2022]
Abstract
Hypoxia-inducible factor 1α (HIF1α) has emerged as a promising new target for pancreatic cancer treatment over the past decade. High expression of HIF-1α increases the drug resistance of the current first line chemotherapeutic drug, gemcitabine (Gem). Here we employed biocompatible lipid-polymer hybrid nanoparticles to co-deliver HIF1α siRNA (si-HIF1α) and Gem for pancreatic cancer treatment in subcutaneous and orthotopic tumor models. The cationic ε-polylysine co-polymer (ENPs) can effectively absorb negatively charged si-HIF1α on the surface and encapsulate Gem to the hydrophilic core. Further coating of ENPs with PEGylated lipid bilayer resulted formation of LENPs, with reversed surface charge. The lipid bilayer of LENPs prevented nanoparticle aggregation and si-HIF1α degradation in serum, as well as Gem leakage. Those characteristics endow LENPs encapsulating drug prolonged lifetime in bloodstream and improved drug release via the enhanced tumor vasculature effect in tumor tissues. LENPs can co-deliver Gem and si-HIF1α (LENP-Gem-si-HIF1α) into tumor cells and effectively suppress the HIF1α expression both in vitro and in vivo. LENP-Gem-siHIF1α exhibited significant synergistic antitumor effects. Furthermore, LENP-Gem-si-HIF1α showed excellent capability to inhibit tumor metastasis in orthotopic tumor model.
Collapse
Affiliation(s)
- Xiao Zhao
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology (NCNST), 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Feng Li
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology (NCNST), 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Yiye Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology (NCNST), 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology (NCNST), 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - He Ren
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Jing Chen
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology (NCNST), 11 Beiyitiao, Zhongguancun, Beijing 100190, China.
| | - Jihui Hao
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.
| |
Collapse
|
93
|
Lytton-Jean AKR, Kauffman KJ, Kaczmarek JC, Langer R. Cancer nanotherapeutics in clinical trials. Cancer Treat Res 2015; 166:293-322. [PMID: 25895874 DOI: 10.1007/978-3-319-16555-4_13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
To be legally sold in the United States, all drugs must go through the FDA approval process. This chapter introduces the FDA approval process and describes the clinical trials required for a drug to gain approval. We then look at the different cancer nanotherapeutics and in vivo diagnostics that are currently in clinical trials or have already received approval. These nanotechnologies are catagorized and described based on the delivery vehicle: liposomes, polymer micelles, albumin-bound chemotherapeutics, polymer-bound chemotherapeutics, and inorganic particles.
Collapse
Affiliation(s)
- Abigail K R Lytton-Jean
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | | | | | | |
Collapse
|
94
|
Chu D, Xu W, Pan R, Chen P. Co-delivery of drug nanoparticles and siRNA mediated by a modified cell penetrating peptide for inhibiting cancer cell proliferation. RSC Adv 2015. [DOI: 10.1039/c4ra14827d] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Modified cell penetrating peptide can stabilize drug nanoparticles with improved efficacy and co-deliver siRNA inducing synergy on the inhibition of cancer cell growth.
Collapse
Affiliation(s)
- Dafeng Chu
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology
- University of Waterloo
- Waterloo
- Canada
| | - Wen Xu
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology
- University of Waterloo
- Waterloo
- Canada
| | - Ran Pan
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology
- University of Waterloo
- Waterloo
- Canada
| | - P. Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology
- University of Waterloo
- Waterloo
- Canada
| |
Collapse
|
95
|
Zhu K, Lai H, Guo C, Li J, Wang Y, Wang L, Wang C. Nanovector-based prolyl hydroxylase domain 2 silencing system enhances the efficiency of stem cell transplantation for infarcted myocardium repair. Int J Nanomedicine 2014; 9:5203-15. [PMID: 25429216 PMCID: PMC4243506 DOI: 10.2147/ijn.s71586] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has attracted much attention in myocardial infarction therapy. One of the limitations is the poor survival of grafted cells in the ischemic microenvironment. Small interfering RNA-mediated prolyl hydroxylase domain protein 2 (PHD2) silencing in MSCs holds tremendous potential to enhance their survival and paracrine effect after transplantation. However, an efficient and biocompatible PHD2 silencing system for clinical application is lacking. Herein, we developed a novel PHD2 silencing system based on arginine-terminated generation 4 poly(amidoamine) (Arg-G4) nanoparticles. The system exhibited effective and biocompatible small interfering RNA delivery and PHD2 silencing in MSCs in vitro. After genetically modified MSC transplantation in myocardial infarction models, MSC survival and paracrine function of IGF-1 were enhanced significantly in vivo. As a result, we observed decreased cardiomyocyte apoptosis, scar size, and interstitial fibrosis, and increased angiogenesis in the diseased myocardium, which ultimately attenuated ventricular remodeling and improved heart function. This work demonstrated that an Arg-G4 nanovector-based PHD2 silencing system could enhance the efficiency of MSC transplantation for infarcted myocardium repair.
Collapse
Affiliation(s)
- Kai Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| | - Changfa Guo
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| | - Jun Li
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| | - Yulin Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| | - Lingyan Wang
- Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| |
Collapse
|
96
|
Sun T, Zhang YS, Pang B, Hyun DC, Yang M, Xia Y. Engineered nanoparticles for drug delivery in cancer therapy. Angew Chem Int Ed Engl 2014; 53:12320-64. [PMID: 25294565 DOI: 10.1002/anie.201403036] [Citation(s) in RCA: 744] [Impact Index Per Article: 74.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Indexed: 12/18/2022]
Abstract
In medicine, nanotechnology has sparked a rapidly growing interest as it promises to solve a number of issues associated with conventional therapeutic agents, including their poor water solubility (at least, for most anticancer drugs), lack of targeting capability, nonspecific distribution, systemic toxicity, and low therapeutic index. Over the past several decades, remarkable progress has been made in the development and application of engineered nanoparticles to treat cancer more effectively. For example, therapeutic agents have been integrated with nanoparticles engineered with optimal sizes, shapes, and surface properties to increase their solubility, prolong their circulation half-life, improve their biodistribution, and reduce their immunogenicity. Nanoparticles and their payloads have also been favorably delivered into tumors by taking advantage of the pathophysiological conditions, such as the enhanced permeability and retention effect, and the spatial variations in the pH value. Additionally, targeting ligands (e.g., small organic molecules, peptides, antibodies, and nucleic acids) have been added to the surface of nanoparticles to specifically target cancerous cells through selective binding to the receptors overexpressed on their surface. Furthermore, it has been demonstrated that multiple types of therapeutic drugs and/or diagnostic agents (e.g., contrast agents) could be delivered through the same carrier to enable combination therapy with a potential to overcome multidrug resistance, and real-time readout on the treatment efficacy. It is anticipated that precisely engineered nanoparticles will emerge as the next-generation platform for cancer therapy and many other biomedical applications.
Collapse
Affiliation(s)
- Tianmeng Sun
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332 (USA)
| | | | | | | | | | | |
Collapse
|
97
|
Sun T, Zhang YS, Pang B, Hyun DC, Yang M, Xia Y. Maßgeschneiderte Nanopartikel für den Wirkstofftransport in der Krebstherapie. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201403036] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
98
|
Hu TY. Multidisciplinary efforts driving translational theranostics. Am J Cancer Res 2014; 4:1209-10. [PMID: 25285169 PMCID: PMC4183998 DOI: 10.7150/thno.10503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 09/08/2014] [Indexed: 02/06/2023] Open
Abstract
This themed issue summarizes significant efforts aimed at using "biological language" to discern between "friends" and "foes" in the context of theranostics for true clinical application. It is expected that the success of theranostics depends on multidisciplinary efforts, combined to expedite our understanding of host responses to "customized" theranostic agents and formulating individualized therapies.
Collapse
|
99
|
Chu D, Xu W, Pan R, Ding Y, Sui W, Chen P. Rational modification of oligoarginine for highly efficient siRNA delivery: structure-activity relationship and mechanism of intracellular trafficking of siRNA. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 11:435-46. [PMID: 25193363 DOI: 10.1016/j.nano.2014.08.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/17/2014] [Accepted: 08/21/2014] [Indexed: 11/29/2022]
Abstract
UNLABELLED Recently, cell-penetrating peptides (CPPs) have received much attention for cellular delivery of therapeutic molecules. However, in the case of CPPs as carriers for siRNA delivery, their utility is often restricted by low cellular uptake and/or entrapment in endosomes. Here, in order to deliver siRNAs with high efficiency, oligoarginine, a prominent member in CPPs, is rationally modified with oligohistidine and stearyl moieties (STR-) by fully taking into account the formation of nanoparticles, uptake and intracellular trafficking. We show that when the ratio of histidine/arginine in a peptide sequence is >1.5, pronounced gene silencing is induced. Following this rule, STR-HnR8 (n=16 and 20) are developed, which show a high knockdown efficiency rarely reported before. Finally, we find that endosomal escape of siRNA induced by stearylated and oligohistidylated oligoarginine is only from "proton-sponge" effect. Taken together, our results suggest a new strategy for the improvement of CPP-based siRNA delivery systems. FROM THE CLINICAL EDITOR This study present a novel cell penetrating peptide-based siRNA delivery system utilizing modified oligo-arginine demonstrating a successful siRNA delivery approach.
Collapse
Affiliation(s)
- Dafeng Chu
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario, Canada
| | - Wen Xu
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario, Canada
| | - Ran Pan
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario, Canada
| | - Yong Ding
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario, Canada
| | - Weiping Sui
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario, Canada
| | - P Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario, Canada.
| |
Collapse
|
100
|
Liu X, Liu C, Catapano CV, Peng L, Zhou J, Rocchi P. Structurally flexible triethanolamine-core poly(amidoamine) dendrimers as effective nanovectors to deliver RNAi-based therapeutics. Biotechnol Adv 2014; 32:844-52. [DOI: 10.1016/j.biotechadv.2013.08.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 08/01/2013] [Indexed: 12/31/2022]
|