1
|
Patil PJ, Sutar SS, Usman M, Patil DN, Dhanavade MJ, Shehzad Q, Mehmood A, Shah H, Teng C, Zhang C, Li X. Exploring bioactive peptides as potential therapeutic and biotechnology treasures: A contemporary perspective. Life Sci 2022; 301:120637. [PMID: 35568229 DOI: 10.1016/j.lfs.2022.120637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/02/2022] [Accepted: 05/09/2022] [Indexed: 12/21/2022]
Abstract
In preceding years, bioactive peptides (BAPs) have piqued escalating attention owing to their multitudinous biological features. To date, many potential BAPs exhibiting anti-cancer activities have been documented; yet, obstacles such as their safety profiles and consumer acceptance continue to exist. Moreover, BAPs have been discovered to facilitate the suppression of Coronavirus Disease 2019 (CoVID-19) and maybe ideal for treating the CoVID-19 infection, as stated by published experimental findings, but their widespread knowledge is scarce. Likewise, there is a cornucopia of BAPs possessing neuroprotective effects that mend neurodegenerative diseases (NDs) by regulating gut microbiota, but they remain a subject of research interest. Additionally, a plethora of researchers have attempted next-generation approaches based on BAPs, but they need scientific attention. The text format of this critical review is organized around an overview of BAPs' versatility and diverse bio functionalities with emphasis on recent developments and novelties. The review is alienated into independent sections, which are related to either BAPs based disease management strategies or next-generation BAPs based approaches. BAPs based anti-cancer, anti-CoVID-19, and neuroprotective strategies have been explored, which may offer insights that could help the researchers and industries to find an alternate regimen against the three aforementioned fatal diseases. To the best of our knowledge, this is the first review that has systematically discussed the next-generation approaches in BAP research. Furthermore, it can be concluded that the BAPs may be optimal for the management of cancer, CoVID-19, and NDs; nevertheless, experimental and preclinical studies are crucial to validate their therapeutic benefits.
Collapse
Affiliation(s)
- Prasanna J Patil
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China; School of Food and Health, Beijing Technology and Business University, No. 11, Fucheng Road, Beijing 100048, China
| | - Shubham S Sutar
- Department of Biotechnology, Shivaji University, Vidyanagar, Kolhapur, Maharashtra 416004, India
| | - Muhammad Usman
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China; School of Food and Health, Beijing Technology and Business University, No. 11, Fucheng Road, Beijing 100048, China
| | - Devashree N Patil
- Department of Biotechnology, Shivaji University, Vidyanagar, Kolhapur, Maharashtra 416004, India
| | - Maruti J Dhanavade
- Department of Microbiology, Bharati Vidyapeeth's Dr. Patangrao Kadam Mahavidyalaya, Sangli, Maharashtra 416416, India
| | - Qayyum Shehzad
- National Engineering Laboratory for Agri-Product Quality Traceability, Beijing Technology and Business University, Beijing 100048, China
| | - Arshad Mehmood
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China; School of Food and Health, Beijing Technology and Business University, No. 11, Fucheng Road, Beijing 100048, China; Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing 100048, China
| | - Haroon Shah
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China; School of Food and Health, Beijing Technology and Business University, No. 11, Fucheng Road, Beijing 100048, China
| | - Chao Teng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China; School of Food and Health, Beijing Technology and Business University, No. 11, Fucheng Road, Beijing 100048, China; Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing 100048, China
| | - Chengnan Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China; School of Food and Health, Beijing Technology and Business University, No. 11, Fucheng Road, Beijing 100048, China; Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing 100048, China.
| | - Xiuting Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China; School of Food and Health, Beijing Technology and Business University, No. 11, Fucheng Road, Beijing 100048, China; Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing 100048, China.
| |
Collapse
|
2
|
Wang J, Chen G, Liu N, Han X, Zhao F, Zhang L, Chen P. Strategies for improving the safety and RNAi efficacy of noncovalent peptide/siRNA nanocomplexes. Adv Colloid Interface Sci 2022; 302:102638. [PMID: 35299136 DOI: 10.1016/j.cis.2022.102638] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 12/12/2022]
Abstract
In the past decades, the striking development of cationic polypeptides and cell-penetrating peptides (CPPs) tailored for small interfering RNA (siRNA) delivery has been fuelled by the conception of nuclear acid therapy and precision medicine. Owing to their amino acid compositions, inherent secondary structures as well as diverse geometrical shapes, peptides or peptide-containing polymers exhibit good biodegradability, high flexibility, and bio-functional diversity as nonviral siRNA vectors. Also, a variety of noncovalent nanocomplexes could be built via self-assembling and electrostatic interactions between cationic peptides and siRNAs. Although the peptide/siRNA nanocomplex-based RNAi therapies, STP705 and MIR-19, are under clinical trials, a guideline addressing the current bottlenecks of peptide/siRNA nanocomplex delivery is in high demand for future research and development. In this review, we present strategies for improving the safety and RNAi efficacy of noncovalent peptide/siRNA nanocomplexes in the treatment of genetic disorders. Through thorough analysis of those RNAi formulations using different delivery strategies, we seek to shed light on the rationale of peptide design and modification in constructing robust siRNA delivery systems, including targeted and co-delivery systems. Based on this, we provide a timely and comprehensive understanding of how to engineer biocompatible and efficient peptide-based siRNA vectors.
Collapse
Affiliation(s)
- Jun Wang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Guang Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada; Key Laboratory of Chemical Additives for China National Light Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Nan Liu
- Advanced Materials Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250014, China
| | - Xiaoxia Han
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Feng Zhao
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - P Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada; Advanced Materials Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250014, China.
| |
Collapse
|
3
|
Ryu YC, Lee YE, Hwang BH. Efficient and safe small RNA delivery to macrophage using peptide-based nanocomplex. Biotechnol Bioeng 2021; 119:482-492. [PMID: 34761810 DOI: 10.1002/bit.27988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/30/2021] [Accepted: 11/01/2021] [Indexed: 01/29/2023]
Abstract
As one of the gene therapies, RNA interference (RNAi) effectively suppresses only specific genes, targeting various diseases in which they are involved. For the successful process of RNAi, efficient and safe delivery of small RNAs, including small interfering RNA and short hairpin RNA, is essential. Herein, an S-R11 fusion peptide, SPACE peptide conjugated with poly-arginine, was introduced to deliver small RNAs into immune cells that are difficult to transfect. This S-R11 peptide stably formed a spontaneous self-assembling nanocomplex through electrostatic attraction and hydrogen bonding with small RNAs. The nanocomplex showed about 5.3-fold better permeation efficiency than the conventional Lipofectamine™ 2000 for RAW 264.7 macrophage cells. Moreover, it induced about 66.2% silencing effect of the target gene in the cells activated with polyinosinic:polycytidylic acid (poly (I:C)). In addition, the cell viability of fusion peptide was ensured even in a concentration range exceeding the concentration used in the nanocomplex. Based on these results, it is expected that the nanocomplex in this study can be used as a new gene delivery system that can overcome the challenge of gene therapies to immune cells.
Collapse
Affiliation(s)
- Yeong Chae Ryu
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Incheon, Korea
| | - Yoo Eun Lee
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Incheon, Korea
| | - Byeong Hee Hwang
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Incheon, Korea.,Division of Bioengineering, Incheon National University, Incheon, Korea
| |
Collapse
|
4
|
Ryu YC, Kim KA, Kim BC, Wang HMD, Hwang BH. Novel fusion peptide-mediated siRNA delivery using self-assembled nanocomplex. J Nanobiotechnology 2021; 19:44. [PMID: 33579303 PMCID: PMC7881583 DOI: 10.1186/s12951-021-00791-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
Background Gene silencing using siRNA can be a new potent strategy to treat many incurable diseases at the genetic level, including cancer and viral infections. Treatments using siRNA essentially requires an efficient and safe method of delivering siRNA into cells while maintaining its stability. Thus, we designed novel synergistic fusion peptides, i.e., SPACE and oligoarginine. Results Among the novel fusion peptides and siRNAs, nanocomplexes have enhanced cellular uptake and gene silencing effect in vitro and improved retention and gene silencing effects of siRNAs in vivo. Oligoarginine could attract siRNAs electrostatically to form stable and self-assembled nanocomplexes, and the SPACE peptide could interact with the cellular membrane via hydrogen bonding. Therefore, nanocomplexes using fusion peptides showed improved and evident cellular uptake and gene silencing of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) via the lipid raft-mediated endocytosis pathway, especially to the HDFn cells of the skin, and all of the fusion peptides were biocompatible. Also, intratumorally injected nanocomplexes had increased retention time of siRNAs at the site of the tumor. Finally, nanocomplexes demonstrated significant in vivo gene silencing effect without overt tissue damage and immune cell infiltration. Conclusions The new nanocomplex strategy could become a safe and efficient platform for the delivery of siRNAs into cells and tissues to treat various target diseases through gene silencing.![]()
Collapse
Affiliation(s)
- Yeong Chae Ryu
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Academy-ro 119, Yeonsu-gu, Incheon, 22012, Korea
| | - Kyung Ah Kim
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Academy-ro 119, Yeonsu-gu, Incheon, 22012, Korea
| | - Byoung Choul Kim
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Academy-ro 119, Yeonsu-gu, Incheon, 22012, Korea.,Division of Bioengineering, Incheon National University, Incheon, 22012, Korea
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, 402, Taiwan
| | - Byeong Hee Hwang
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Academy-ro 119, Yeonsu-gu, Incheon, 22012, Korea. .,Division of Bioengineering, Incheon National University, Incheon, 22012, Korea.
| |
Collapse
|
5
|
Chopra M, Sgro A, Norret M, Blancafort P, Iyer KS, Evans CW. A peptide-functionalised dendronised polymer for selective transfection in human liver cancer cells. NEW J CHEM 2021. [DOI: 10.1039/d1nj01566d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A dendronised polymer functionalised with SP94 targeting peptide achieves highly selective transient transfection of liver cancer cells over normal non-transformed hepatocytes.
Collapse
Affiliation(s)
- Meenu Chopra
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Agustin Sgro
- The Harry Perkins Institute of Medical Research, 6 Verdun St, Nedlands, WA 6009, Australia
- School of Human Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Marck Norret
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Pilar Blancafort
- The Harry Perkins Institute of Medical Research, 6 Verdun St, Nedlands, WA 6009, Australia
- School of Human Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - K. Swaminathan Iyer
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Cameron W. Evans
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| |
Collapse
|
6
|
Dutta K, Bochicchio D, Ribbe AE, Alfandari D, Mager J, Pavan GM, Thayumanavan S. Symbiotic Self-Assembly Strategy toward Lipid-Encased Cross-Linked Polymer Nanoparticles for Efficient Gene Silencing. ACS APPLIED MATERIALS & INTERFACES 2019; 11:24971-24983. [PMID: 31264399 DOI: 10.1021/acsami.9b04731] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
A novel "symbiotic self-assembly" strategy that integrates the advantages of biocompatible lipids with a structurally robust polymer to efficiently encapsulate and deliver siRNAs is reported. The assembly process is considered to be symbiotic because none of the assembling components are capable of self-assembly but can form well-defined nanostructures in the presence of others. The conditions of the self-assembly process are simple but have been chosen such that it offers the ability to arrive at a system that is noncationic for mitigating carrier-based cytotoxicity, efficiently encapsulate siRNA to minimize cargo loss, be effectively camouflaged to protect the siRNA from nuclease degradation, and efficiently escape the endosome to cause gene knockdown. The lipid-siRNA-polymer (L-siP) nanoassembly formation and its disassembly in the presence of an intracellular trigger have been extensively characterized experimentally and through computational modeling. The complexes have been evaluated for the delivery of four different siRNA molecules in six different cell lines, where an efficient gene knockdown is demonstrated. The reported generalized strategy has the potential to make an impact on the development of a safe and effective delivery agent for RNAi-mediated gene therapy that holds the promise of targeting several hard-to-cure diseases.
Collapse
Affiliation(s)
| | - Davide Bochicchio
- Department of Innovative Technologies , University of Applied Sciences and Arts of Southern Switzerland , CH-6928 Manno , Switzerland
| | | | | | | | - Giovanni M Pavan
- Department of Innovative Technologies , University of Applied Sciences and Arts of Southern Switzerland , CH-6928 Manno , Switzerland
- Department of Applied Science and Technology , Politecnico di Torino , Corso Duca degli Abruzzi 24 , 10129 Torino , Italy
| | | |
Collapse
|
7
|
Shuai Q, Zhao G, Zhang X, Yu B, Lee RJ, Su WK. Selection of fluorescent dye for tracking biodistribution of paclitaxel in live imaging. Colloids Surf B Biointerfaces 2019; 181:872-878. [PMID: 31382335 DOI: 10.1016/j.colsurfb.2019.06.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/14/2019] [Accepted: 06/16/2019] [Indexed: 11/17/2022]
Abstract
Fluorescence imaging is widely used to determine biodistribution of drugs in mice. However, the dye distribution may not be able to exactly reflect the true distribution of drug molecules. We synthesized PTX-Cy5.5 and mPEG-PLA-Cy5.5, and then prepared dye-loaded nanoparticles (NPs) (Cy5.5, DiR, PTX-Cy5.5, and mPEG-PLA-Cy5.5), dye and PTX co-loaded NPs, and PTX-loaded NPs, respectively. The particle sizes of resulting NPs were between 42.7 nm and 68.8 nm, and Zeta potential was between -0.86 mV and -8.49 mV. The biodistribution of fluorescent NPs (dye-loaded NPs and dye and PTX co-loaded NPs) on Bel-7402 tumor-bearing mice was studied via in vivo fluorescence imaging assays, results of which suggested that Cy5.5 loaded NPs and Cy5.5 conjugates (PTX-Cy5.5 and mPEG-PLA-Cy5.5) formulated NPs can reflect the tissue distribution of PTX whether it was incorporated or not. However, DiR failed to reflect true tissue distribution of PTX unless it was co-loaded with PTX. Based on these results, a guidance for the selection of dyes in drug distribution investigations and disease-targeted treatment was presented.
Collapse
Affiliation(s)
- Qi Shuai
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Guangkuo Zhao
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xiaomin Zhang
- Hangzhou Push-Kang Biotechnology Co., Ltd, Zhejiang, China
| | - Bo Yu
- Hangzhou Push-Kang Biotechnology Co., Ltd, Zhejiang, China
| | - Robert J Lee
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Wei-Ke Su
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
8
|
Zhao Y, Lee RJ, Liu L, Dong S, Zhang J, Zhang Y, Yao Y, Lu J, Meng Q, Xie J, Teng L. Multifunctional drug carrier based on PEI derivatives loaded with small interfering RNA for therapy of liver cancer. Int J Pharm 2019; 564:214-224. [PMID: 31004717 DOI: 10.1016/j.ijpharm.2019.04.049] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/18/2019] [Accepted: 04/16/2019] [Indexed: 02/08/2023]
Abstract
Gene therapy strategies for liver cancer have broad application prospects but still lack a stable and efficient delivery vehicle. To overcome this obstacle, we designed a multifunctional gene delivery vector, sTPssOLP, which was based on oleylamine (OA)-modified disulfide-containing polyethylenimine (PEI) and incorporated into lipids to prepare a lipid nanoparticle. sTPssOLP consisted of the core of PEI derivative and cationic lipids bound to siRNA. The modified polyethylene glycol (PEG) and transferrin (Tf) were partially embedded in the phospholipid bilayer through the lipid and the other as the outer shell. The aim was to use the redox responsiveness of disulfide to trigger siRNA release in cytoplasm to enhance transfection efficiency. Pegylated lipids and Tf focus on increasing cycle life in the body and increasing accumulation at the tumor site of the carrier. In addition, two vectors were prepared as controls, one based on a PEI derivative containing no disulfide bond (POLP) and the other on the surface of the carrier not linked to Tf (PssOLP). PEI derivatives effectively avoid the toxicity problems caused by the use of PEI alone (25 kDa). Meanwhile, it was confirmed by gel retardation experiments that in the presence of dithiothreitol (DTT), the disulfide bond can indeed be reduced and the siRNA entrapped in the vector can be released. Both HepG2 and SMMC had significant uptake of sTPssOLP. The results of intracellular and lysosomal co-localization indicated that sTPssOLP achieved lysosomal escape. RT-PCR and Western blot results also confirmed that sTPssOLP had the best gene silencing activity. In vivo, the tumor inhibition rate of sTPssOLP in nude mice carrying HepG2 xenografts was 56%, which was significantly greater than that of the saline control group. In vivo imaging results showed that fluorescently labeled siRNA loaded in sTPssOLP was able to deliver more to the tumor site. At the same time, it was observed that sTPssOLP did not show significant damage to normal tissues. Therefore, this multifunctional gene delivery vector warrants further investigation.
Collapse
Affiliation(s)
- Yarong Zhao
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Robert J Lee
- Jilin University, School of Life Sciences, Changchun, Jilin, China; Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Luotong Liu
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Shiyan Dong
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Jing Zhang
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Yu Zhang
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | | | - Jiahui Lu
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Qingfan Meng
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Jing Xie
- Jilin University, School of Life Sciences, Changchun, Jilin, China.
| | - Lesheng Teng
- Jilin University, School of Life Sciences, Changchun, Jilin, China.
| |
Collapse
|
9
|
Wang X, Xiao X, Zhang B, Li J, Zhang Y. A self-assembled peptide nucleic acid-microRNA nanocomplex for dual modulation of cancer-related microRNAs. Chem Commun (Camb) 2019; 55:2106-2109. [PMID: 30698603 DOI: 10.1039/c9cc00002j] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Here, we report a new strategy for the construction of a peptide nucleic acid-microRNA nanocomplex with dual function to simultaneously suppress oncogenic microRNAs and upregulate tumor-suppressive microRNAs in target cancer cells.
Collapse
Affiliation(s)
- Xingxing Wang
- State Key Laboratory of Analytical Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | | | | | | | | |
Collapse
|
10
|
Xie K, Song S, Zhou L, Wan J, Qiao Y, Wang M, Xie H, Zhou L, Zheng S, Wang H. Revival of a potent therapeutic maytansinoid agent using a strategy that combines covalent drug conjugation with sequential nanoparticle assembly. Int J Pharm 2018; 556:159-171. [PMID: 30553007 DOI: 10.1016/j.ijpharm.2018.12.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/10/2018] [Accepted: 12/03/2018] [Indexed: 01/16/2023]
Abstract
Maytansine and its related analogues are a class of highly potent anti-proliferation agents that have failed to be exploited as clinical drugs for human therapy due to unacceptable systemic toxicity. Here, we delineate a novel strategy that combines rational drug conjugation with subsequent nanoparticle assembly to systemically deliver this highly potent and toxic drug. To demonstrate this concept, we covalently coupled the thiolated maytansine derivative, the DM1 agent, to amphiphilic block co-polymers, polyethylene glycol (PEG)-block-polylactide (PLA), in varying molecular weights to generate two prodrug constructs (i.e., PEG2K-PLA2K-DM1 and PEG2K-PLA4K-DM1) via the maleimide-thiol reaction. The resulting two constructs are amenable to self-assembly in aqueous solutions and are systemically injectable for preclinical studies. In vivo evaluations indicate that PEG-PLA-DM1 conjugate-assembled nanoparticles (NPs) display substantially reduced drug toxicity compared to the free drug forms and NPs that physically encapsulate DM1. Furthermore, following systemic administration, these nanodrugs produced superior therapeutic efficacy over free DM1 in a colon tumor xenograft-bearing mouse model. Therefore, this study provides evidence that the conjugation of toxic drugs to assembling copolymers enables the alleviation of cancer drug toxicity and effective delivery of anticancer drugs. Thus, this DM1-formulated platform represents a new generation of nanotherapeutics that are available for further clinical evaluation.
Collapse
Affiliation(s)
- Ke Xie
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Shanshan Song
- Institute of Microanalytical Systems, Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China
| | - Liqian Zhou
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Jianqin Wan
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Yiting Qiao
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Min Wang
- Institute of Microanalytical Systems, Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China
| | - Haiyang Xie
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Lin Zhou
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Shusen Zheng
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China.
| | - Hangxiang Wang
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China.
| |
Collapse
|
11
|
Chen W, Zhou Y, Zhi X, Ma T, Liu H, Chen BW, Zheng X, Xie S, Zhao B, Feng X, Dang X, Liang T. Delivery of miR-212 by chimeric peptide-condensed supramolecular nanoparticles enhances the sensitivity of pancreatic ductal adenocarcinoma to doxorubicin. Biomaterials 2018; 192:590-600. [PMID: 30553134 DOI: 10.1016/j.biomaterials.2018.11.035] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 11/15/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a destructive cancer with poor prognosis. Both novel therapeutic targets and approaches are needed to improve the overall survival of PDAC patients. MicroRNA-212 (miR-212) has been reported as a tumor suppressor in multiple cancers, but its definitive role and exact mechanism in the progression of pancreatic cancer is unclear. In this study, we developed a new chimeric peptide (PL-1) composed of plectin-1-targeted PDAC-specific and arginine-rich RNA-binding motifs which could condense miRNA to self-assemble supramolecular nanoparticles. These nanoparticles could deliver miR-212 into PDAC cells specifically and efficiently which also showed good stability in RNase and serum. Moreover, we demonstrated that PL-1/miR-212 nanoparticles could dramatically enhance the chemotherapeutic effect of doxorubicin for PDAC both in vitro and in vivo. In terms of mechanism, combined miR-212 intervention by PL-1/miR-212 nanoparticles resulted in obvious decrease of USP9X expression (ubiquitin specific peptidase 9, X-linked, USP9X) and eventually enhanced the doxorubicin induced apoptosis and autophagy of PDAC cells. These findings provide a new promising anti-cancer strategy via PL-1/miR-212 nanoparticles and identify miR-212/USP9X as a new potential target for future systemic therapy against human PDAC.
Collapse
Affiliation(s)
- Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Zhi
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Ma
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Liu
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Brayant Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxiao Zheng
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shangzhi Xie
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Zhao
- Life Sciences Institute, Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Xinhua Feng
- Life Sciences Institute, Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Xiaowei Dang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China.
| |
Collapse
|
12
|
Wang C, Chen L, Huang Y, Li K, Jinye A, Fan T, Zhao R, Xia X, Shen B, Du J, Liu Y. Exosome-delivered TRPP2 siRNA inhibits the epithelial-mesenchymal transition of FaDu cells. Oncol Lett 2018; 17:1953-1961. [PMID: 30675260 DOI: 10.3892/ol.2018.9752] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 10/23/2018] [Indexed: 12/14/2022] Open
Abstract
The prognosis for patients with head and neck cancer (HNC) remains poor, owing to uncontrolled tumor invasion and metastasis. Epithelial-mesenchymal transition (EMT) serves an important role in this invasion and metastasis, and transient receptor potential polycystic 2 (TRPP2) enhances metastasis and invasion by regulating EMT in human laryngeal squamous cell carcinoma. The present study examined whether exosomes/TRPP2 small interfering RNA (siRNA) complexes were able to reduce EMT by suppressing TRPP2 expression in FaDu cells, a cell line of human pharyngeal squamous cell carcinoma. Using agarose gel electrophoresis, it was determined that exosome/TRPP2 siRNA complexes were stable in the presence of nucleases and serum. A fluorescence assay and western blotting analysis was performed, and it was reported that the FaDu cells took up exosomes, the exosomes effectively delivered TRPP2 siRNA into FaDu cells and that exosome/TRPP2 siRNA complexes significantly suppressed TRPP2 protein expression levels in FaDu cells. Furthermore, expression levels of E-cadherin were significantly increased, whereas expression levels of N-cadherin and vimentin were significantly decreased in FaDu cells transfected with TRPP2 siRNA. Thus, exosome/TRPP2 siRNA complexes markedly suppressed TRPP2 expression and EMT in FaDu cells. These results suggested that further development of exosome/TRPP2 siRNA complexes for use as an RNA-based gene therapy in the treatment of HNC is warranted.
Collapse
Affiliation(s)
- Chunhui Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Lei Chen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yuanyuan Huang
- Department of Gastroenterology and Hepatology, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Kun Li
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Anqi Jinye
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Taotao Fan
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Ren Zhao
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xianming Xia
- Department of Gastroenterology and Hepatology, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Bing Shen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Juan Du
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yehai Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
13
|
Song Z, Chen X, You X, Huang K, Dhinakar A, Gu Z, Wu J. Self-assembly of peptide amphiphiles for drug delivery: the role of peptide primary and secondary structures. Biomater Sci 2018; 5:2369-2380. [PMID: 29051950 DOI: 10.1039/c7bm00730b] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Peptide amphiphiles (PAs), functionalized with alkyl chains, are capable of self-assembling into various nanostructures. Recently, PAs have been considered as ideal drug carriers due to their good biocompatibility, specific biological functions, and hypotoxicity to normal cells and tissues. Meanwhile, the nanocarriers formed by PAs are able to achieve controlled drug release and enhanced cell uptake in response to the stimulus of the physiological environment or specific biological factors in the location of the lesion. However, the underlying detailed drug delivery mechanism, especially from the aspect of primary and secondary structures of PAs, has not been systematically summarized or discussed. Focusing on the relationship between the primary and secondary structures of PAs and stimuli-responsive drug delivery applications, this review highlights the recent advances, challenges, and opportunities of PA-based functional drug nanocarriers, and their potential pharmaceutical applications are discussed.
Collapse
Affiliation(s)
- Zhenhua Song
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Engineering, Sun Yat-sen University, Guangzhou, 510006, PR China.
| | | | | | | | | | | | | |
Collapse
|
14
|
Wang H, Zhou L, Xie K, Wu J, Song P, Xie H, Zhou L, Liu J, Xu X, Shen Y, Zheng S. Polylactide-tethered prodrugs in polymeric nanoparticles as reliable nanomedicines for the efficient eradication of patient-derived hepatocellular carcinoma. Theranostics 2018; 8:3949-3963. [PMID: 30083272 PMCID: PMC6071539 DOI: 10.7150/thno.26161] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/02/2018] [Indexed: 12/26/2022] Open
Abstract
Nanomedicines have been extensively explored for cancer treatment, and their efficacies have arguably been proven in various cancer cell-derived xenograft (CDX) mouse models. However, they generally fail to show such therapeutic advantages in patients because of the huge pathological differences between human tumors and CDX models. Methods: In this study, we fabricated colloidal ultrastable nanomedicines from polymeric prodrugs and compared the therapeutic efficacies in hepatocellular carcinoma (HCC) CDX and clinically relevant patient-derived xenograft (PDX) mouse models, which closely mimic human tumor pathological properties. Working towards this goal, we esterified a highly potent SN38 (7-ethyl-10-hydroxycamptothecin) agent using oligo- or polylactide (oLA or PLA) segments with varying molecular weights. Results: The resulting SN38 conjugates assembled with polyethylene glycol-block-polylactic acid to form systemically injectable nanomedicines. With increasing PLA chain length, the SN38 conjugates showed extended retention in the nanoparticles and superior antitumor activity, completely eradicating xenografted tumors in both mouse models. Our data implicate that these small-sized and ultrastable nanomedicines might also efficaciously treat cancer in patients. More interestingly, the systemically delivered nanomedicines notably alleviated the incidence of bloody diarrhea. Conclusion: Our studies demonstrate that the appropriate molecular editing of anticancer drugs enables the generation of better tolerated cytotoxic nanotherapy for cancer, which represents a potentially useful scaffold for further clinical translation.
Collapse
Affiliation(s)
- Hangxiang Wang
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine; Zhejiang University, Hangzhou 310003, P. R. China
- Shenzhen Key Laboratory of Hepatobiliary Disease, Shenzhen Third People's Hospital, Shenzhen 518112, P. R. China
| | - Liqian Zhou
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine; Zhejiang University, Hangzhou 310003, P. R. China
| | - Ke Xie
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine; Zhejiang University, Hangzhou 310003, P. R. China
| | - Jiaping Wu
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine; Zhejiang University, Hangzhou 310003, P. R. China
| | - Penghong Song
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine; Zhejiang University, Hangzhou 310003, P. R. China
| | - Haiyang Xie
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine; Zhejiang University, Hangzhou 310003, P. R. China
| | - Lin Zhou
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine; Zhejiang University, Hangzhou 310003, P. R. China
| | - Jialin Liu
- Shenzhen Key Laboratory of Hepatobiliary Disease, Shenzhen Third People's Hospital, Shenzhen 518112, P. R. China
| | - Xiao Xu
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine; Zhejiang University, Hangzhou 310003, P. R. China
| | - Youqing Shen
- Center for Bionanoengineering and State Key Laboratory of Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Shusen Zheng
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine; Zhejiang University, Hangzhou 310003, P. R. China
| |
Collapse
|
15
|
A nanomedicine approach enables co-delivery of cyclosporin A and gefitinib to potentiate the therapeutic efficacy in drug-resistant lung cancer. Signal Transduct Target Ther 2018; 3:16. [PMID: 29942660 PMCID: PMC6013461 DOI: 10.1038/s41392-018-0019-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/26/2018] [Accepted: 05/15/2018] [Indexed: 12/11/2022] Open
Abstract
Drug resistance, accounting for therapeutic failure in the clinic, remains a major challenge to effectively manage cancer. Cyclosporin A (CsA) can reverse multidrug resistance (MDR), especially resistance to epidermal growth factor receptor tyrosine kinase inhibitors. However, the application of both drugs in cancer therapies is hampered by their poor aqueous solubility and low bioavailability due to oral administration. CsA augments the potency of gefitinib (Gef) in both Gef-sensitive and Gef-resistant cell lines. Here, we show that the simultaneous encapsulation of CsA and Gef within polyethylene glycol-block-poly(D, L-lactic acid) (PEG-PLA) produced a stable and systemically injectable nanomedicine, which exhibited a sub-50-nm diameter and spherical structures. Impressively, the co-delivery of therapeutics via single nanoparticles (NPs) outperformed the oral administration of the free drug combination at suppressing tumor growth. Furthermore, in vivo results indicated that CsA formulated in NPs sensitized Gef-resistant cells and Gef-resistant tumors to Gef treatment by inactivating the STAT3/Bcl-2 signaling pathway. Collectively, our nanomedicine approach not only provides an alternative administration route for the drugs of choice but also effectively reverses MDR, facilitating the development of effective therapeutic modalities for cancer. Injection of nanoparticles containing the anticancer drug gefitinib and the immunosuppressant cyclosporin A reverses drug-resistant cancer growth in mice. The development of multidrug resistance is the main reason why many forms of chemotherapy fail. Cyclosporin A, a drug used to prevent immune rejection after organ transplantation, has previously been shown to enhance the potency of gefitinib. Hangxiang Wang and colleagues at Zhejiang University, Hangzhou, China, have successfully combined cyclosporin A and gefitinib, two poorly water-soluble and slowly absorbed drugs, into stable injectable nanoparticles that delay the growth of gefitinib resistant human lung cancer cells as well as the growth of drug resistant tumors in mice. Importantly, this novel co-formulation was more effective than oral co-administration of the two drugs. Further investigation into this drug delivery route could yield much needed alternative treatments for patients with multidrug-resistant cancers.
Collapse
|
16
|
Abstract
Self-assembled peptide nanostructures have been increasingly exploited as functional materials for applications in biomedicine and energy. The emergent properties of these nanomaterials determine the applications for which they can be exploited. It has recently been appreciated that nanomaterials composed of multicomponent coassembled peptides often display unique emergent properties that have the potential to dramatically expand the functional utility of peptide-based materials. This review presents recent efforts in the development of multicomponent peptide assemblies. The discussion includes multicomponent assemblies derived from short low molecular weight peptides, peptide amphiphiles, coiled coil peptides, collagen, and β-sheet peptides. The design, structure, emergent properties, and applications for these multicomponent assemblies are presented in order to illustrate the potential of these formulations as sophisticated next-generation bio-inspired materials.
Collapse
Affiliation(s)
- Danielle M Raymond
- Department of Chemistry, University of Rochester, Rochester, NY 14627-0216, USA.
| | | |
Collapse
|
17
|
Spicer CD, Jumeaux C, Gupta B, Stevens MM. Peptide and protein nanoparticle conjugates: versatile platforms for biomedical applications. Chem Soc Rev 2018; 47:3574-3620. [PMID: 29479622 PMCID: PMC6386136 DOI: 10.1039/c7cs00877e] [Citation(s) in RCA: 283] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Peptide- and protein-nanoparticle conjugates have emerged as powerful tools for biomedical applications, enabling the treatment, diagnosis, and prevention of disease. In this review, we focus on the key roles played by peptides and proteins in improving, controlling, and defining the performance of nanotechnologies. Within this framework, we provide a comprehensive overview of the key sequences and structures utilised to provide biological and physical stability to nano-constructs, direct particles to their target and influence their cellular and tissue distribution, induce and control biological responses, and form polypeptide self-assembled nanoparticles. In doing so, we highlight the great advances made by the field, as well as the challenges still faced in achieving the clinical translation of peptide- and protein-functionalised nano-drug delivery vehicles, imaging species, and active therapeutics.
Collapse
Affiliation(s)
- Christopher D Spicer
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, Stockholm, Sweden.
| | | | | | | |
Collapse
|
18
|
Qiao Y, Wan J, Zhou L, Ma W, Yang Y, Luo W, Yu Z, Wang H. Stimuli‐responsive nanotherapeutics for precision drug delivery and cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 11:e1527. [DOI: 10.1002/wnan.1527] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/16/2018] [Accepted: 03/23/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Yiting Qiao
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi‐Organ Transplantation, Ministry of Public Health, School of Medicine Zhejiang University Hangzhou P.R. China
| | - Jianqin Wan
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi‐Organ Transplantation, Ministry of Public Health, School of Medicine Zhejiang University Hangzhou P.R. China
- Department of Chemical Engineering Zhejiang University Hangzhou P.R. China
| | - Liqian Zhou
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi‐Organ Transplantation, Ministry of Public Health, School of Medicine Zhejiang University Hangzhou P.R. China
| | - Wen Ma
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening Southern Medical University Guangzhou P.R. China
| | - Yuanyuan Yang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening Southern Medical University Guangzhou P.R. China
| | - Weixuan Luo
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening Southern Medical University Guangzhou P.R. China
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening Southern Medical University Guangzhou P.R. China
| | - Hangxiang Wang
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi‐Organ Transplantation, Ministry of Public Health, School of Medicine Zhejiang University Hangzhou P.R. China
| |
Collapse
|
19
|
Guan X, Chang Y, Sun J, Song J, Xie Y. Engineered Hsp Protein Nanocages for siRNA Delivery. Macromol Biosci 2018; 18:e1800013. [DOI: 10.1002/mabi.201800013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/24/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Xingang Guan
- Life Science Research Center; Beihua University; Jilin 132013 P. R. China
| | - Yu Chang
- Life Science Research Center; Beihua University; Jilin 132013 P. R. China
- College of Medicine; Beihua University; Jilin 132013 P. R. China
| | - Jinghui Sun
- College of Medicine; Beihua University; Jilin 132013 P. R. China
| | - Jianxi Song
- College of Medicine; Beihua University; Jilin 132013 P. R. China
| | - Yu Xie
- Life Science Research Center; Beihua University; Jilin 132013 P. R. China
| |
Collapse
|
20
|
The Hippo pathway as a drug target in gastric cancer. Cancer Lett 2018; 420:14-25. [PMID: 29408652 DOI: 10.1016/j.canlet.2018.01.062] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 02/08/2023]
Abstract
The Hippo tumor suppressor pathway is critical for balancing cellular differentiation and proliferation in response to cell-cell contact, mechanical signals and diffusible signals such as lysophosphatidic acid. Hippo pathway signaling is frequently dysregulated in gastric cancer (GC), as well as many other kinds of solid tumors, contributing to multiple aspects of malignant progression including unchecked cell division and metastasis. Considering the importance of this Hippo pathway in cancer, its pharmacological disruption may be of huge benefit in the fight against this disease. In this review, we summarize the components of the Hippo pathway, its crosstalk with other major oncogenic signaling pathways, common mechanisms of its dysregulation, as well as potential therapeutic approaches of targeting this pathway for cancer treatment, specifically in a GC context.
Collapse
|
21
|
Xu L, Xu S, Wang H, Zhang J, Chen Z, Pan L, Wang J, Wei X, Xie H, Zhou L, Zheng S, Xu X. Enhancing the Efficacy and Safety of Doxorubicin against Hepatocellular Carcinoma through a Modular Assembly Approach: The Combination of Polymeric Prodrug Design, Nanoparticle Encapsulation, and Cancer Cell-Specific Drug Targeting. ACS APPLIED MATERIALS & INTERFACES 2018; 10:3229-3240. [PMID: 29313660 DOI: 10.1021/acsami.7b14496] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Intervention is urgently required to improve the therapeutic outcome for patients with unresectable hepatocellular carcinomas (HCCs). However, current chemotherapeutics, such as sorafenib and doxorubicin (DOX), provide only limited therapeutic benefits for this devastating disease. In this context, we present a modular assembly approach to the construction of a systemically injectable nanotherapeutic that can efficiently and safely deliver DOX in vivo. To achieve this goal, we covalently attached DOX to a polylactide (PLA) building block (Mw = 2800, n = 36), yielding DOX-PLA conjugate 1. Due to the lipophilicity imparted by PLA, the conjugate 1 coassembled with an amphiphilic lipid, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethylene glycol) 2000] (DSPE-PEG2000), to form nanoparticles (NPs). To achieve preferential tumor accumulation, we additionally decorated the particle surface with an HCC-specific peptide moiety (i.e., SP94). The resulting HCC-targetable DOX-encapsulating NPs (termed tNP-PLA-DOX) exhibited several unique characteristics, including the feasible fabrication of sub-100 nm NPs, substantially delayed drug release profiles of several weeks, HCC cell-specific uptake and tumor accumulation in an in vivo mouse model, as well as alleviated drug toxicity in animals. Collectively, these results show that the integration of multiple components within a single nanocarrier via modular assembly is cost-effective for the creation of safe anticancer nanotherapeutics. The presented DOX-based nanomedicines have potential for enhancing the therapeutic index in patients.
Collapse
Affiliation(s)
- Li Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Shengjun Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Hangxiang Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Jun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Zun Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Linhui Pan
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Jianguo Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Haiyang Xie
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Lin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Shusen Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| |
Collapse
|
22
|
|
23
|
Huang X, Lee RJ, Qi Y, Li Y, Lu J, Meng Q, Teng L, Xie J. Microfluidic hydrodynamic focusing synthesis of polymer-lipid nanoparticles for siRNA delivery. Oncotarget 2017; 8:96826-96836. [PMID: 29228574 PMCID: PMC5722526 DOI: 10.18632/oncotarget.18281] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/01/2017] [Indexed: 12/12/2022] Open
Abstract
Small interfering RNAs (siRNAs) are promising as therapeutics for intractable diseases such as cancer. However, efficient and safe delivery of siRNAs in vivo remains a challenge. Polymer-lipid hybrid nanoparticles (P/LNPs) have been evaluated for therapeutic delivery of siRNA. In this study, a microfluidic hydrodynamic focusing (MF) system was used to prepare P/LNPs loaded with VEGF siRNA. P/LNPs made by MF were smaller in particle size and had narrower size distribution compared to P/LNPs formed by bulk mixing (BM). MF-synthesized P/LNPs demonstrated low vehicle cytotoxicity and potent tumor cell inhibition in vitro. In addition, P/LNPs produced by the microfluidic chip exhibited prolonged blood circulation and increased AUC after i.v. injection compared to free siRNA. Furthermore, P/LNPs synthesized by MF induced greater down-regulation of VEGF mRNA and protein levels as well as greater tumor inhibition in a xenograft tumor model. Taken together, P/LNPs prepared by MF have been shown to be an effective and safe therapeutic siRNA delivery system for cancer treatment both in vitro and in vivo.
Collapse
Affiliation(s)
- Xueqin Huang
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China.,Department of Chemistry and Pharmacy, Zhuhai College of Jilin University, Zhuhai, Guangdong, 519041, China
| | - Robert J Lee
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China.,Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA
| | - Yuhang Qi
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| | - Yujing Li
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| | - Jiahui Lu
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| | - Qingfan Meng
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| | - Jing Xie
- School of Life Sciences, Jilin University, Changchun, Jilin 130023, China
| |
Collapse
|
24
|
Wang H, Wu J, Xie K, Fang T, Chen C, Xie H, Zhou L, Zheng S. Precise Engineering of Prodrug Cocktails into Single Polymeric Nanoparticles for Combination Cancer Therapy: Extended and Sequentially Controllable Drug Release. ACS APPLIED MATERIALS & INTERFACES 2017; 9:10567-10576. [PMID: 28271714 DOI: 10.1021/acsami.7b01938] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The synergistic combination of two or more chemotherapeutics frequently requires packaging in single delivery vehicles for the sequential release of each substance in a predictable manner. Here, we demonstrate for the first time that the rational engineering of a prodrug cocktail into single polymeric nanoparticles (NPs) can enable the sequential release of chemotherapeutics in a controllable manner. Exploiting combretastatin-A4 (CA4, 1) as a model antiangiogenesis agent, two ester derivatives, 2 and 3, tethered with saturated fatty acids (butanoic and heptanoic acid for 2 and 3, respectively) were synthesized. 7-Ethyl-10-hydroxycamptothecin (SN38) derivative 4, esterified with α-linolenic acid, was used as a cytotoxic drug. Because of their augmented lipophilicity and miscibility, all constructed prodrugs readily assembled with clinically approved polymeric matrices. Results showed that altering the aliphatic chains of modifiers for CA4 chemical derivatization enabled the drug retention capacity within particle systems to be adjusted, leading to the identification of the prodrug cocktail of 2 and 4 as an optimal combination for subsequent preclinical studies. Furthermore, in vivo assessements indicated that the resulting NPs simultaneously formulating 2 and 4 exhibited synergistic activities and outperformed NPs loaded with individual prodrugs 2 or 4 in terms of therapeutic efficacy. These findings highlight a novel and versatile strategy for tailoring chemically disparate prodrug cocktails for adaptation within a single nanoplatform as a potential modality for synergistic cancer therapy.
Collapse
Affiliation(s)
- Hangxiang Wang
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Jiaping Wu
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Ke Xie
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Tao Fang
- Jinhua People's Hospital , Jinhua, Zhejiang Province 321000, P.R. China
| | - Chao Chen
- College of Life Sciences, Huzhou University , Huzhou, Zhejiang Province 313000, P.R. China
| | - Haiyang Xie
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Lin Zhou
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Shusen Zheng
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| |
Collapse
|
25
|
Yuan Y, Xu L, Dai S, Wang M, Wang H. A facile supramolecular approach to fabricate multifunctional upconversion nanoparticles as a versatile platform for drug loading, in vivo delivery and tumor imaging. J Mater Chem B 2017; 5:2425-2435. [PMID: 32264550 DOI: 10.1039/c6tb03381d] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Multifunctional upconversion nanoparticles (UCNPs) that can be implemented in theranostic applications are particularly attractive scaffolds for precise drug delivery. However, most of the current methods for drug formulation are technically complicated, thereby impeding their use in the clinic. Here, we report on the preparation of a lipophilic cytotoxic prodrug-integrated and polyethylene glycol (PEG)-cloaked UCNPs scaffold through a facile one-pot supramolecular approach. By choosing 7-ethyl-10-hydroxycamptothecin (SN38)-derived prodrug 1 as a model chemotherapeutic, we show that this lipophilic prodrug can be feasibly self-assembled onto the surface of UCNPs, which are cooperatively solubilized by PEGylated phospholipids. The resulting SN38 prodrug 1-encapsulated UCNPs (designated 1@pUCNPs) produce a stable colloidal system in aqueous solution, making it suitable for intravenous injection. The SN38 drug loading capacity in pUCNPs is as high as ∼12.3 wt%, and a sustained drug release profile is observed, indicating that the drug payloads can be transported to targeted tumor sites via the enhanced permeability and retention (EPR) effect. Upconversion luminescence (UCL) imaging, including in vivo and ex vivo imaging, suggests that the drug-loaded pUCNPs remain stable in tumors over a long time and preferentially accumulate in tumors presumably via the EPR effect. Furthermore, the 1@pUCNPs show superior therapeutic outcomes compared with the clinically approved SN38 prodrug CPT-11 in the Bcap-37 mouse model of breast cancer. Collectively, our results demonstrate that pUCNPs facilely constructed in a one-pot self-assembly manner may be used as a versatile platform, enabling synchronous in vivo delivery of poorly water-soluble drugs and tumor imaging.
Collapse
Affiliation(s)
- Yingying Yuan
- The First Affiliated Hospital, School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University, Hangzhou 310003, P. R. China.
| | | | | | | | | |
Collapse
|
26
|
Yin F, Hu K, Chen S, Wang D, Zhang J, Xie M, Yang D, Qiu M, Zhang H, Li ZG. Black phosphorus quantum dot based novel siRNA delivery systems in human pluripotent teratoma PA-1 cells. J Mater Chem B 2017; 5:5433-5440. [DOI: 10.1039/c7tb01068k] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
As a novel semiconducting materials, BP-QDs possess superior transfection efficiency, excellent biocompatibility and low cytotoxicity, which shows promising potential for siRNA delivery and photothermal effects in cancer therapy.
Collapse
Affiliation(s)
- Feng Yin
- School of Chemical Biology and Biotechnology
- Peking University Shenzhen Graduate School
- Shenzhen
- China
| | - Kuan Hu
- School of Chemical Biology and Biotechnology
- Peking University Shenzhen Graduate School
- Shenzhen
- China
| | - Si Chen
- SZU-NUS Collaborative Innovation Center for Optoelectronic Science and Technology and Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province College of Optoelectronic Engineering
- Shenzhen University
- Shenzhen 518060
- P. R. China
| | - Dongyuan Wang
- School of Chemical Biology and Biotechnology
- Peking University Shenzhen Graduate School
- Shenzhen
- China
| | - Jianing Zhang
- School of Chemical Biology and Biotechnology
- Peking University Shenzhen Graduate School
- Shenzhen
- China
| | - Mingsheng Xie
- School of Chemical Biology and Biotechnology
- Peking University Shenzhen Graduate School
- Shenzhen
- China
| | - Dan Yang
- School of Chemical Biology and Biotechnology
- Peking University Shenzhen Graduate School
- Shenzhen
- China
| | - Meng Qiu
- SZU-NUS Collaborative Innovation Center for Optoelectronic Science and Technology and Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province College of Optoelectronic Engineering
- Shenzhen University
- Shenzhen 518060
- P. R. China
| | - Han Zhang
- SZU-NUS Collaborative Innovation Center for Optoelectronic Science and Technology and Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province College of Optoelectronic Engineering
- Shenzhen University
- Shenzhen 518060
- P. R. China
| | - Zi-gang Li
- School of Chemical Biology and Biotechnology
- Peking University Shenzhen Graduate School
- Shenzhen
- China
| |
Collapse
|
27
|
Integrating a novel SN38 prodrug into the PEGylated liposomal system as a robust platform for efficient cancer therapy in solid tumors. Int J Pharm 2016; 512:39-48. [PMID: 27544846 DOI: 10.1016/j.ijpharm.2016.08.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 07/26/2016] [Accepted: 08/17/2016] [Indexed: 12/20/2022]
Abstract
Liposomal nanoassemblies have been used extensively as carriers for the delivery of both lipophilic and hydrophilic drugs. They represent a mature, versatile technology with considerable potential for improving the pharmacokinetics of drugs. However, the formulation of many chemotherapeutics into liposome systems has posed a significant challenge due to their incompatible physicochemical properties, as was the case with camptothecin-based chemotherapeutics. Here, we present a rational paradigm of potent chemotherapeutics that were reconstructed and subsequently integrated into liposomal nanoassemblies. Using SN38 (7-ethyl-10-hydroxy camptothecin) as a model drug, a lipophilic prodrug 1 (designated as LA-SN38) was constructed by tethering the linoleic acid (LA) moiety via esterification, which was further facilitated to form liposomal nanoparticles (LipoNP) through supramolecular nanoassembly. The resulting 1-loaded LipoNP exhibited sustained drug release kinetics and decreased cellular uptake by macrophage cells. Uptake by tumor cells was enhanced relative to our previous supramolecular nanoparticles (SNP 1), which were derived from the self-assembling prodrug 1. Notably, LipoNP outperformed SNP 1 in terms of pharmacokinetics and in vivo therapeutic efficacy in both human BEL-7402 hepatocellular carcinoma (HCC) and HCT-116 colorectal cancer-derived xenograft mouse models. These results were likely due to the improved systemic circulation and preferential accumulation of nanodrugs in tumors. Hence, our results suggest that the combination of liposomal delivery platforms with rational prodrug engineering may emerge as a promising approach for the effective and safe delivery of anticancer chemotherapeutics.
Collapse
|
28
|
Han Y, Li H, Hu Y, Li P, Wang H, Nie Z, Yao S. Time-Resolved Luminescence Biosensor for Continuous Activity Detection of Protein Acetylation-Related Enzymes Based on DNA-Sensitized Terbium(III) Probes. Anal Chem 2015; 87:9179-85. [DOI: 10.1021/acs.analchem.5b01338] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Yitao Han
- State Key Laboratory of Chemo/Biosensing
and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People’s Republic of China
| | - Hao Li
- State Key Laboratory of Chemo/Biosensing
and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People’s Republic of China
| | - Yufang Hu
- State Key Laboratory of Chemo/Biosensing
and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People’s Republic of China
| | - Pei Li
- State Key Laboratory of Chemo/Biosensing
and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People’s Republic of China
| | - Huixia Wang
- State Key Laboratory of Chemo/Biosensing
and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People’s Republic of China
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing
and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People’s Republic of China
| | - Shouzhuo Yao
- State Key Laboratory of Chemo/Biosensing
and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People’s Republic of China
| |
Collapse
|
29
|
Pan D, Sun J, Jin H, Li Y, Li L, Wu Y, Zhang L, Yang Z. Supramolecular assemblies of novel aminonucleoside phospholipids and their bonding to nucleic acids. Chem Commun (Camb) 2015; 51:469-72. [PMID: 25383905 DOI: 10.1039/c4cc07538b] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A novel class of aminonucleoside phospholipids has been developed. These molecules could spontaneously assemble into supramolecular structures including multilamellar organization, hydrogels, superhelical strands, and vesicles. Their ability to bind to DNA by hydrogen bonding and π-π stacking interactions was investigated by many means.
Collapse
Affiliation(s)
- Delin Pan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road 38, Beijing 100191, China.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Gao H, Takemoto H, Chen Q, Naito M, Uchida H, Liu X, Miyata K, Kataoka K. Regulated protonation of polyaspartamide derivatives bearing repeated aminoethylene side chains for efficient intracellular siRNA delivery with minimal cytotoxicity. Chem Commun (Camb) 2015; 51:3158-61. [DOI: 10.1039/c4cc09859e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The protonation behavior of aminoethylene units in polyaspartamide side-chains was modulated for improving the delivery efficiency of siRNA-loaded polyion complexes.
Collapse
Affiliation(s)
- Hui Gao
- School of Chemistry and Chemical Engineering
- Tianjin University of Technology
- Tianjin
- China
- Department of Materials Engineering
| | - Hiroyasu Takemoto
- Polymer Chemistry Division
- Chemical Resources Laboratory
- Tokyo Institute of Technology
- Yokohama 226-8503
- Japan
| | - Qixian Chen
- Department of Materials Engineering
- Graduate School of Engineering
- The University of Tokyo
- Tokyo 113-8656
- Japan
| | - Mitsuru Naito
- Department of Materials Engineering
- Graduate School of Engineering
- The University of Tokyo
- Tokyo 113-8656
- Japan
| | - Hirokuni Uchida
- Center for Disease Biology and Integrative Medicine
- Graduate School of Medicine
- The University of Tokyo
- Tokyo 113-0033
- Japan
| | - Xueying Liu
- Center for Disease Biology and Integrative Medicine
- Graduate School of Medicine
- The University of Tokyo
- Tokyo 113-0033
- Japan
| | - Kanjiro Miyata
- Center for Disease Biology and Integrative Medicine
- Graduate School of Medicine
- The University of Tokyo
- Tokyo 113-0033
- Japan
| | - Kazunori Kataoka
- Department of Materials Engineering
- Graduate School of Engineering
- The University of Tokyo
- Tokyo 113-8656
- Japan
| |
Collapse
|