1
|
Zhang J, Liu S, Wang Y, Li X, Zeng H, Li B, Wang J. Preparation of Chitosan Nanoparticles through a Readily Solvent-Exchange Process for Efficient and Enhanced Gene Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:10486-10491. [PMID: 38728233 DOI: 10.1021/acs.langmuir.3c03874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
In view of the excellent prospects of gene therapy and the potential safety and immunogenicity issues challenged by viral vectors, it is of great significance to develop a nonviral vector with low toxicity and low cost. In this work, we report a chitosan nanoparticle (CSNP) to be used as a gene vector prepared through a facile solvent-exchange strategy. Chitosan is first dissolved in ionic liquid 1-ethyl-3-methylimidazolium acetate (EMIM Ac), and then, the solvent is exchanged with water/phosphate-buffered saline (PBS) to remove ionic liquid, forming a final CSNP dispersion after ultrasonication. The prepared CSNP shows a positive surface charge and can condense green fluorescent protein-encoding plasmid (pGFP) at weight ratios (CSNP/pGFP) of 5/1 or higher. Dynamic light scattering size and ζ-potential characterization and gel retardation results confirm the formation of CSNP/pGFP complexes. Compared with plain pGFP, efficient cellular internalization and significantly enhanced green fluorescent protein (GFP) expression are observed by using CSNP as a plasmid vector. Benefitting from the intrinsic biocompatibility, low cost, low immunogenicity, and abundant sources of chitosan, as well as the facile preparation and the efficient gene transfection capacity of CSNP, it is believed that this CSNP could be used as a nonviral gene vector with great clinical translational potentials.
Collapse
Affiliation(s)
- Jialuo Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Shujing Liu
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yulin Wang
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiaoxu Li
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Huazhang Zeng
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Boxuan Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| |
Collapse
|
2
|
Zhang J, Xiao Y, Zhang J, Yang Y, Zhang L, Liang F. Recent advances of engineered oncolytic viruses-based combination therapy for liver cancer. J Transl Med 2024; 22:3. [PMID: 38167076 PMCID: PMC10763442 DOI: 10.1186/s12967-023-04817-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
Liver cancer is a major malignant tumor, which seriously threatens human health and increases the economic burden on patients. At present, gene therapy has been comprehensively studied as an excellent therapeutic measure in liver cancer treatment. Oncolytic virus (OV) is a kind of virus that can specifically infect and kill tumor cells. After being modified by genetic engineering, the specificity of OV infection to tumor cells is increased, and its influence on normal cells is reduced. To date, OV has shown its effectiveness and safety in experimental and clinical studies on a variety of tumors. Thus, this review primarily introduces the current status of different genetically engineered OVs used in gene therapy for liver cancer, focuses on the application of OVs and different target genes for current liver cancer therapy, and identifies the problems encountered in OVs-based combination therapy and the corresponding solutions, which will provide new insights into the treatment of liver cancer.
Collapse
Affiliation(s)
- Junhe Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China.
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China.
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Yunxi Xiao
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
| | - Jie Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yun Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liao Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
| | - Fan Liang
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
| |
Collapse
|
3
|
Ma Y, Li S, Lin X, Chen Y. Bioinspired Spatiotemporal Management toward RNA Therapies. ACS NANO 2023; 17:24539-24563. [PMID: 38091941 DOI: 10.1021/acsnano.3c08219] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Ribonucleic acid (RNA)-based therapies have become an attractive topic in disease intervention, especially with some that have been approved by the FDA such as the mRNA COVID-19 vaccine (Comirnaty, Pfizer-BioNTech, and Spikevax, Moderna) and Patisiran (siRNA-based drug for liver delivery). However, extensive applications are still facing challenges in delivering highly negatively charged RNA to the targeted site. Therapeutic delivery strategies including RNA modifications, RNA conjugates, and RNA polyplexes and delivery platforms such as viral vectors, nanoparticle-based delivery platforms, and hydrogel-based delivery platforms as potential nucleic acid-releasing depots have been developed to enhance their cellular uptake and protect nucleic acid from being degraded by immune systems. Here, we review the growing number of viral vectors, nanoparticles, and hydrogel-based RNA delivery systems; describe RNA loading/release mechanism induced by environmental stimulations including light, heat, pH, or enzyme; discuss their physical or chemical interactions; and summarize the RNA therapeutics release period (temporal) and their target cells/organs (spatial). Finally, we describe current concerns, highlight current challenges and future perspectives of RNA-based delivery systems, and provide some possible research areas that provide opportunities for clinical translation of RNA delivery carriers.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Shiyao Li
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Xin Lin
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27705, United States
| | - Yupeng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
4
|
Guo X, Lin J, Pan L, He K, Huang Z, Chen J, Lin C, Zeng B, Luo S, Wang M. Ultrasound-triggered release of miR-199a-3p from liposome nanobubbles for enhanced hepatocellular carcinoma treatment. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:560-571. [PMID: 37850395 DOI: 10.1080/21691401.2023.2268137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023]
Abstract
This study was aimed to develop an efficient tumour-targeted liposome nanobubbles (LNBs) system using ultrasound-targeted nanobubble destruction for enhanced release and transfection of miRNA-199a-3p in hepatocellular carcinoma (HCC) therapy. The prepared LNBs comprised a polyethylene glycol-modified liposome shell and a perfluoropentane (PFP) core. MiRNA-199a-3p was attached to the nanocomposite surface via electrostatic adsorption, while RGD peptide functionalized the LNBs surface for enhanced HCC cell targeting, namely PFP@miR-RGD-LNBs. The LNBs were spherical with a narrow size distribution. The gene-loaded LNBs effectively condensed miR-199a-3p and protected it from enzymatic degradation. Low-intensity focused ultrasound (LIFU) promoted a fast release of miR-199a-3p from the prepared LNBs, thereby enhancing therapeutic effects. The combined application of PFP@miR-RGD-LNBs and LIFU exhibited a more potent inhibitory effect on HepG2 cells than the other groups, potentially due to LIFU promoting rapid and efficient gene release at the target site and increasing cell membrane permeability. Quantitative reverse transcription-polymerase chain reaction analysis revealed significantly increased mRNA expression levels of key apoptosis markers (Bad, Bax, Caspase-9 and Caspase-3) in the PFP@miR-RGD-LNBs + LIFU group compared to other groups. These findings suggest that the prepared LNBs are highly likely to be promising candidates for further exploration of HCC gene delivery and therapy.
Collapse
Affiliation(s)
- Xinmin Guo
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Jianru Lin
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Liwen Pan
- Department of Endocrinology, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Kun He
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Zhihui Huang
- Department of Nuclear Medicine, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Jialin Chen
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Cuiyan Lin
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Baohui Zeng
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Sijia Luo
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Mengdie Wang
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
5
|
Bowman KER, Ahne L, O'Brien L, Vander Mause ER, Lu P, Wallis B, Evason KJ, Lim CS. p53-Bad* Fusion Gene Therapy Induces Apoptosis In Vitro and Reduces Zebrafish Tumor Burden in Hepatocellular Carcinoma. Mol Pharm 2023; 20:331-340. [PMID: 36490361 PMCID: PMC10760808 DOI: 10.1021/acs.molpharmaceut.2c00665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
With few curative treatments and a global yearly death rate of over 800,000, hepatocellular carcinoma (HCC) desperately needs new therapies. Although wild-type p53 gene therapy has been shown to be safe in HCC patients, it has not shown enough efficacy to merit approval. This work aims to show how p53 can be re-engineered through fusion to the pro-apoptotic BH3 protein Bcl-2 antagonist of cell death (Bad) to improve anti-HCC activity and potentially lead to a novel HCC therapeutic, p53-Bad*. p53-Bad* is a fusion of p53 and Bad, with two mutations, S112A and S136A. We determined mitochondrial localization of p53-Bad* in liver cancer cell lines with varying p53 mutation statuses via fluorescence microscopy. We defined the apoptotic activity of p53-Bad* in four liver cancer cell lines using flow cytometry. To determine the effects of p53-Bad* in vivo, we generated and analyzed transgenic zebrafish expressing hepatocyte-specific p53-Bad*. p53-Bad* localized to the mitochondria regardless of the p53 mutation status and demonstrated superior apoptotic activity over WT p53 in early, middle, and late apoptosis assays. Tumor burden in zebrafish HCC was reduced by p53-Bad* as measured by the liver-to-body mass ratio and histopathology. p53-Bad* induced significant apoptosis in zebrafish HCC as measured by TUNEL staining but did not induce apoptosis in non-HCC fish. p53-Bad* can induce apoptosis in a panel of liver cancer cell lines with varying p53 mutation statuses and induce apoptosis/reduce HCC tumor burden in vivo in zebrafish. p53-Bad* warrants further investigation as a potential new HCC therapeutic.
Collapse
Affiliation(s)
- Katherine E Redd Bowman
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Lisa Ahne
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
- Institute of Pharmacy, Experimental Pharmacology for Natural Sciences, Martin Luther University, Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Liam O'Brien
- Department of Pathology and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, United States
| | - Erica R Vander Mause
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Phong Lu
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Bryce Wallis
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Kimberley J Evason
- Department of Pathology and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, United States
| | - Carol S Lim
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
6
|
Virosome, a promising delivery vehicle for siRNA delivery and its novel preparation method. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
7
|
Huang L, Jiang Y, Liu X, Guo Y, Feng Y, Pan P, Li M, Liu Y. Antheraea pernyi silk fibroin-coated adenovirus as a VEGF165-Ang-1 dual gene delivery vector. J BIOACT COMPAT POL 2022. [DOI: 10.1177/08839115221095254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Vascularization is a key challenge in the regeneration of tissues containing blood vessels. In this study, spermine was used for cationic modification of Antheraea pernyi silk fibroin (ASF) to synthesize cationized ASF (CASF). CASF/Ad complexes prepared by coating adenovirus (Ad) with CASF were used as delivery vectors for vascular endothelial growth factor 165 and angiopoietin-1 dual genes. The results showed that the zeta potential of the Ad was reversed from −7.75 mV to approximately +8.40 mV after CASF coating, and the sizes of the CASF/Ad complexes were 200 to 290 nm. Furthermore, human umbilical vein endothelial cells HUVECs were cocultured and infected with CASF/Ad in vitro. The results of confocal laser scanning microscopy, flow cytometry and CCK-8 assay showed that coating Ad with CASF at concentration of 20 and 50 µg/mL not only reduced the cytotoxicity of naked Ad, but also significantly promoted cell proliferation. Therefore, the CASF/Ad complexes could be beneficial to reduce the dosage of Ad and the potential toxicity risk of high doses of Ad in vivo, which has the potential of application to promote vascular network regeneration.
Collapse
Affiliation(s)
- Linling Huang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Yi Jiang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Xueping Liu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Ying Guo
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Yanfei Feng
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Peng Pan
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Mingzhong Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Yu Liu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| |
Collapse
|
8
|
Xi S, Yang YG, Suo J, Sun T. Research Progress on Gene Editing Based on Nano-Drug Delivery Vectors for Tumor Therapy. Front Bioeng Biotechnol 2022; 10:873369. [PMID: 35419357 PMCID: PMC8996155 DOI: 10.3389/fbioe.2022.873369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/11/2022] [Indexed: 12/25/2022] Open
Abstract
Malignant tumors pose a serious threat to human health and have high fatality rates. Conventional clinical anti-tumor treatment is mainly based on traditional surgery, chemotherapy, radiotherapy, and interventional therapy, and even though these treatment methods are constantly updated, a satisfactory efficacy is yet to be obtained. Therefore, research on novel cancer treatments is being actively pursued. We review the classification of gene therapies of malignant tumors and their advantages, as well as the development of gene editing techniques. We further reveal the nano-drug delivery carrier effect in improving the efficiency of gene editing. Finally, we summarize the progress in recent years of gene editing techniques based on nano-drug delivery carriers in the treatment of various malignant tumors, and analyze the prospects of the technique and its restricting factors.
Collapse
Affiliation(s)
- Shiwen Xi
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- Gastrointestinal Surgical Department, The First Hospital, Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Jian Suo
- Gastrointestinal Surgical Department, The First Hospital, Jilin University, Changchun, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| |
Collapse
|
9
|
Lan T, Que H, Luo M, Zhao X, Wei X. Genome editing via non-viral delivery platforms: current progress in personalized cancer therapy. Mol Cancer 2022; 21:71. [PMID: 35277177 PMCID: PMC8915502 DOI: 10.1186/s12943-022-01550-8] [Citation(s) in RCA: 19] [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] [Received: 12/14/2021] [Accepted: 02/24/2022] [Indexed: 02/08/2023] Open
Abstract
Cancer is a severe disease that substantially jeopardizes global health. Although considerable efforts have been made to discover effective anti-cancer therapeutics, the cancer incidence and mortality are still growing. The personalized anti-cancer therapies present themselves as a promising solution for the dilemma because they could precisely destroy or fix the cancer targets based on the comprehensive genomic analyses. In addition, genome editing is an ideal way to implement personalized anti-cancer therapy because it allows the direct modification of pro-tumor genes as well as the generation of personalized anti-tumor immune cells. Furthermore, non-viral delivery system could effectively transport genome editing tools (GETs) into the cell nucleus with an appreciable safety profile. In this manuscript, the important attributes and recent progress of GETs will be discussed. Besides, the laboratory and clinical investigations that seek for the possibility of combining non-viral delivery systems with GETs for the treatment of cancer will be assessed in the scope of personalized therapy.
Collapse
Affiliation(s)
- Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Sichuan, 610041, Chengdu, China
| | - Haiying Que
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Sichuan, 610041, Chengdu, China
| | - Min Luo
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Sichuan, 610041, Chengdu, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Sichuan, 610041, Chengdu, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Sichuan, 610041, Chengdu, China.
| |
Collapse
|
10
|
Fu P, Zhang J, Li H, Mak M, Xu W, Tao Z. Extracellular vesicles as delivery systems at nano-/micro-scale. Adv Drug Deliv Rev 2021; 179:113910. [PMID: 34358539 PMCID: PMC8986465 DOI: 10.1016/j.addr.2021.113910] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) have shown significant promises as nano-/micro-size carriers in drug delivery and bioimaging. With more characteristics of EVs explored through tremendous research efforts, their unmatched physicochemical properties, biological features, and mechanical aspects make them unique vehicles, owning exceptional pharmacokinetics, circulatory metabolism and biodistribution pattern when delivering theranostic cargoes. In this review we firstly analyzed pros and cons of the EVs as a delivery platform. Secondly, compared to engineered nanoparticle delivery systems, such as biocompatible di-block co-polymers, rational design to improve EVs (exosomes in particular) were elaborated. Lastly, different pharmaceutical loading approaches into EVs were compared, reaching a conclusion on how to construct a clinically available and effective nano-/micro-carrier for a satisfactory medical mission.
Collapse
Affiliation(s)
- Peiwen Fu
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China; Zhenjiang Municipal Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang 212013, China
| | - Jianguo Zhang
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China; Department of Critical Care Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Haitao Li
- Institute for Energy Research, School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Michael Mak
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University, New Haven 06520, USA.
| | - Wenrong Xu
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China; Zhenjiang Municipal Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang 212013, China.
| | - Zhimin Tao
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China; Zhenjiang Municipal Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang 212013, China.
| |
Collapse
|
11
|
Montaño-Samaniego M, Bravo-Estupiñan DM, Méndez-Guerrero O, Alarcón-Hernández E, Ibáñez-Hernández M. Strategies for Targeting Gene Therapy in Cancer Cells With Tumor-Specific Promoters. Front Oncol 2020; 10:605380. [PMID: 33381459 PMCID: PMC7768042 DOI: 10.3389/fonc.2020.605380] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer is the second cause of death worldwide, surpassed only by cardiovascular diseases, due to the lack of early diagnosis, and high relapse rate after conventional therapies. Chemotherapy inhibits the rapid growth of cancer cells, but it also affects normal cells with fast proliferation rate. Therefore, it is imperative to develop other safe and more effective treatment strategies, such as gene therapy, in order to significantly improve the survival rate and life expectancy of patients with cancer. The aim of gene therapy is to transfect a therapeutic gene into the host cells to express itself and cause a beneficial biological effect. However, the efficacy of the proposed strategies has been insufficient for delivering the full potential of gene therapy in the clinic. The type of delivery vehicle (viral or non viral) chosen depends on the desired specificity of the gene therapy. The first gene therapy trials were performed with therapeutic genes driven by viral promoters such as the CMV promoter, which induces non-specific toxicity in normal cells and tissues, in addition to cancer cells. The use of tumor-specific promoters over-expressed in the tumor, induces specific expression of therapeutic genes in a given tumor, increasing their localized activity. Several cancer- and/or tumor-specific promoters systems have been developed to target cancer cells. This review aims to provide up-to-date information concerning targeting gene therapy with cancer- and/or tumor-specific promoters including cancer suppressor genes, suicide genes, anti-tumor angiogenesis, gene silencing, and gene-editing technology, as well as the type of delivery vehicle employed. Gene therapy can be used to complement traditional therapies to provide more effective treatments.
Collapse
Affiliation(s)
- Mariela Montaño-Samaniego
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Diana M. Bravo-Estupiñan
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Oscar Méndez-Guerrero
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Ernesto Alarcón-Hernández
- Laboratorio de Genética Molecular, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Miguel Ibáñez-Hernández
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
12
|
Kamra M, Maiti B, Saha P, Karande AA, Bhattacharya S. Antibody-Conjugated Vitamin E-Derived Liposomes for Targeted Gene Transfer. ACS APPLIED BIO MATERIALS 2020; 3:8375-8385. [DOI: 10.1021/acsabm.0c00656] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Mohini Kamra
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India
- Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Bappa Maiti
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Pranay Saha
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Anjali A. Karande
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Santanu Bhattacharya
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India
- Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata 700032, India
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| |
Collapse
|
13
|
Yan S, Ren X, Yang J, Wang J, Zhang Q, Xu D. Exosomal miR-548c-5p Regulates Colorectal Cancer Cell Growth and Invasion Through HIF1A/CDC42 Axis. Onco Targets Ther 2020; 13:9875-9885. [PMID: 33116573 PMCID: PMC7547144 DOI: 10.2147/ott.s273008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022] Open
Abstract
Background Mounting evidence has implicated that exosomes-delivered noncoding RNAs are key regulators in carcinogenesis. The effect of miR-548c-5p has been elucidated in some cancers. However, the role of exosomal miR-548c-5p in colorectal cancer (CRC) is not fully understood. We aim to explore the function and mechanism of exosome-delivered miR-548c-5p in CRC. The altering effect of exosome-derived miR-548c-5p on the prognosis of CRC patients is also investigated by estimating overall survival and disease-free survival. Materials and Methods The expression of miR-548c-5p in exosomes is determined by real-time PCR. The proliferation and invasion of CRC cells are estimated by MTT, transwell assay and scratch test. The targeted gene of miR-548c-5p is investigated by luciferase reporter assay, real-time PCR, Western blot and chromosome immunoprecipitation (CHIP) assay. CRC cells are transplanted subcutaneously in BALB/c nude mice to estimate their growth in vivo. Results MiR-548c-5p derived from CRC cell exosomes inhibits the proliferation and invasion of CRC cells in vitro. Exosomal miR-548c-5p can also prevent from colorectal carcinogenesis in nude mice in vivo. HIF1A is documented to be a target of miR-548c-5p, and HIF1A can targetedly regulate CDC42 in CRC cells. Exosomal miR-548c-5p affects CRC cell growth, migration and invasion via miR-548c-5p/HIF1A/CDC42 axis. In addition, exosomal miR-548c-5p can be a predictive factor for CRC prognosis. Conclusion Our study has suggested that exosomal miR-548c-5p can regulate CRC through HIF1A/CDC42 axis, which helps to understand CRC pathogenesis more clearly and identify novel therapeutic strategies for CRC patients.
Collapse
Affiliation(s)
- Shushan Yan
- Department of Gastrointestinal and Anal Diseases Surgery of Affiliated Hospital, Weifang Medical University, Weifang, Shandong Province, People's Republic of China
| | - Xiaoxia Ren
- Department of Gastrointestinal Surgery, Yantai Shan Hospital, Yantai, Shandong Province, People's Republic of China
| | - Jinghan Yang
- Central Laboratory and Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang, Shandong Province, People's Republic of China
| | - Jinghua Wang
- Central Laboratory and Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang, Shandong Province, People's Republic of China
| | - Quan Zhang
- Department of Cardiology of Affiliated Hospital, Weifang Medical University, Weifang, Shandong Province, People's Republic of China
| | - Donghua Xu
- Central Laboratory and Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang, Shandong Province, People's Republic of China
| |
Collapse
|
14
|
Iqubal A, Iqubal MK, Khan A, Ali J, Baboota S, Haque SE. Gene Therapy, A Novel Therapeutic Tool for Neurological Disorders: Current Progress, Challenges and Future Prospective. Curr Gene Ther 2020; 20:184-194. [DOI: 10.2174/1566523220999200716111502] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/02/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
Abstract
:
Neurological disorders are one of the major threat for health care system as they put enormous
socioeconomic burden. All aged populations are susceptible to one or other neurological problems
with symptoms of neuroinflammation, neurodegeneration and cognitive dysfunction. At present,
available pharmacotherapeutics are insufficient to treat these diseased conditions and in most cases,
they provide only palliative effect. It was also found that the molecular etiology of neurological disorders
is directly linked with the alteration in genetic makeup, which can be inherited or triggered by the
injury, environmental toxins and by some existing disease. Therefore, to take care of this situation,
gene therapy has emerged as an advanced modality that claims to permanently cure the disease by deletion,
silencing or edition of faulty genes and by insertion of healthier genes. In this modality, vectors
(viral and non-viral) are used to deliver targeted gene into a specific region of the brain via various
routes. At present, gene therapy has shown positive outcomes in complex neurological disorders, such
as Parkinson's disease, Alzheimer's disease, Huntington disease, Multiple sclerosis, Amyotrophic lateral
sclerosis and in lysosomal storage disease. However, there are some limitations such as immunogenic
reactions non-specificity of viral vectors and a lack of effective biomarkers to understand the efficacy
of therapy. Considerable progress has been made to improve vector design, gene selection and
targeted delivery. This review article deals with the current status of gene therapy in neurological disorders
along with its clinical relevance, challenges and future prospective.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Aamir Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| |
Collapse
|
15
|
Mohammadinejad R, Dehshahri A, Sagar Madamsetty V, Zahmatkeshan M, Tavakol S, Makvandi P, Khorsandi D, Pardakhty A, Ashrafizadeh M, Ghasemipour Afshar E, Zarrabi A. In vivo gene delivery mediated by non-viral vectors for cancer therapy. J Control Release 2020; 325:249-275. [PMID: 32634464 PMCID: PMC7334939 DOI: 10.1016/j.jconrel.2020.06.038] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 12/17/2022]
Abstract
Gene therapy by expression constructs or down-regulation of certain genes has shown great potential for the treatment of various diseases. The wide clinical application of nucleic acid materials dependents on the development of biocompatible gene carriers. There are enormous various compounds widely investigated to be used as non-viral gene carriers including lipids, polymers, carbon materials, and inorganic structures. In this review, we will discuss the recent discoveries on non-viral gene delivery systems. We will also highlight the in vivo gene delivery mediated by non-viral vectors to treat cancer in different tissue and organs including brain, breast, lung, liver, stomach, and prostate. Finally, we will delineate the state-of-the-art and promising perspective of in vivo gene editing using non-viral nano-vectors.
Collapse
Affiliation(s)
- Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Vijay Sagar Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA
| | - Masoumeh Zahmatkeshan
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Pooyan Makvandi
- Institute for Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Naples, Italy; Chemistry Department, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 6153753843, Iran
| | - Danial Khorsandi
- Department of Medical Nanotechnology, Faculty of Advanced, Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran; Department of Biotechnology-Biomedicine, University of Barcelona, Barcelona 08028, Spain
| | - Abbas Pardakhty
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Elham Ghasemipour Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey; Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Turkey.
| |
Collapse
|
16
|
Qu M, Kim HJ, Zhou X, Wang C, Jiang X, Zhu J, Xue Y, Tebon P, Sarabi SA, Ahadian S, Dokmeci MR, Zhu S, Gu Z, Sun W, Khademhosseini A. Biodegradable microneedle patch for transdermal gene delivery. NANOSCALE 2020; 12:16724-16729. [DOI: 10.1039/d0nr02759f] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A gelatin methacryloyl based microneedle patch has been developed for transdermal gene delivery both in vitro and in vivo.
Collapse
|