1
|
Jahanafrooz Z, Oroojalian F, Mokhtarzadeh A, Rahdar A, Díez-Pascual AM. Nanovaccines: Immunogenic tumor antigens, targeted delivery, and combination therapy to enhance cancer immunotherapy. Drug Dev Res 2024; 85:e22244. [PMID: 39138855 DOI: 10.1002/ddr.22244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/16/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024]
Abstract
Nanovaccines have been designed to overcome the limitations associated with conventional vaccines. Effective delivery methods such as engineered carriers or smart nanoparticles (NPs) are critical requisites for inducing self-tolerance and optimizing vaccine immunogenicity with minimum side effects. NPs can be used as adjuvants, immunogens, or nanocarriers to develop nanovaccines for efficient antigen delivery. Multiloaded nanovaccines carrying multiple tumor antigens along with immunostimulants can effectively increase immunity against tumor cells. They can be biologically engineered to boost interactions with dendritic cells and to allow a gradual and constant antigen release. Modifying NPs surface properties, using high-density lipoprotein-mimicking nanodiscs, and developing nano-based artificial antigen-presenting cells such as dendritic cell-derived-exosomes are amongst the new developed technologies to enhance antigen-presentation and immune reactions against tumor cells. The present review provides an overview on the different perspectives, improvements, and barriers of successful clinical application of current cancer therapeutic and vaccination options. The immunomodulatory effects of different types of nanovaccines and the nanoparticles incorporated into their structure are described. The advantages of using nanovaccines to prevent and treat common illnesses such as AIDS, malaria, cancer and tuberculosis are discussed. Further, potential paths to develop optimal cancer vaccines are described. Given the immunosuppressive characteristics of both cancer cells and the tumor microenvironment, applying immunomodulators and immune checkpoint inhibitors in combination with other conventional anticancer therapies are necessary to boost the effectiveness of the immune response.
Collapse
Affiliation(s)
- Zohreh Jahanafrooz
- Department of Biology, Faculty of Sciences, University of Maragheh, Maragheh, Iran
| | - Fatemeh Oroojalian
- Natural Products & Medicinal Plants Research Center, North Khorasan University of Medical Sciences Bojnurd, Bojnurd, Iran
- Department of Medical Nanotechnology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol, Iran
| | - Ana M Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingenieria Química, Alcalá de Henares, Spain
| |
Collapse
|
2
|
Du L, Gong Y, Zhang X, Sun J, Gao F, Shen M, Bai H, Yang T, Cheng X, Li S, Peng J, Liu Z, Ding S, Chen J, Cheng W. PD-L1 siRNA hitched polyethyleneimine-elastase constituting nanovesicle induces tumor immunogenicity and PD-L1 silencing for synergistic antitumor immunotherapy. J Nanobiotechnology 2024; 22:442. [PMID: 39068444 PMCID: PMC11282766 DOI: 10.1186/s12951-024-02700-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND PD-1/PD-L1 blockade has become a powerful method to treat malignant tumors. However, a large proportion of patients still do not benefit from this treatment, due to low tumor immunogenicity and low tumor penetration of the agents. Recently, neutrophil elastase has been shown to induce robust tumor immunogenicity, while the insufficient enzyme activity at the tumor site restricted its anti-tumor application. Here, we designed polyethyleneimine-modified neutrophil elastase (PEI-elastase) loaded with PD-L1small interfering RNA (PD-L1 siRNA) for improving enzymatic activity and delivering siRNA to tumor, which was expected to solve the above-mentioned problems. RESULTS We first demonstrated that PEI-elastase possessed high enzymatic activity, which was also identified as an excellent gene-delivery material. Then, we synthesized anti-tumor lipopolymer (P-E/S Lip) by encapsulating PEI-elastase and PD-L1siRNA with pH-responsive anionic liposomes. The P-E/S Lip could be rapidly cleaved in tumor acidic environment, leading to exposure of the PEI-elastase/PD-L1 siRNA. Consequently, PEI-elastase induced powerful tumor immunogenicity upon direct tumor killing with minimal toxicity to normal cells. In parallel, PEI-elastase delivered PD-L1siRNA into the tumor and reduced PD-L1 expression. Orthotopic tumor administration of P-E/S Lip not only attenuated primary tumor growth, but also produced systemic anti-tumor immune response to inhibit growth of distant tumors and metastasis. Moreover, intravenous administration of P-E/S Lip into mice bearing subcutaneous tumors leaded to an effective inhibition of established B16-F10 tumor and 4T1 tumor, with histological analyses indicating an absence of detectable toxicity. CONCLUSIONS In our study, a protease-based nanoplatform was used to cooperatively provoke robust tumor immunogenicity and down-regulate PD-L1 expression, which exhibited great potential as a combination therapy for precisely treating solid tumors.
Collapse
Affiliation(s)
- Li Du
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Biobank, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yao Gong
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoying Zhang
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jide Sun
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Fengxia Gao
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Meiying Shen
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Huili Bai
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Tiantian Yang
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoxue Cheng
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Siqiao Li
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Jian Peng
- Biobank, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhangling Liu
- Biobank, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Junman Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Wei Cheng
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Biobank, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
3
|
Chen L, Ma Z, Xu C, Xie Y, Ouyang D, Song S, Zhao X, Liu F. Progress in oncolytic viruses modified with nanomaterials for intravenous application. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0275. [PMID: 38009779 PMCID: PMC10690878 DOI: 10.20892/j.issn.2095-3941.2023.0275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/25/2023] [Indexed: 11/29/2023] Open
Abstract
In oncolytic virus (OV) therapy, a critical component of tumor immunotherapy, viruses selectively infect, replicate within, and eventually destroy tumor cells. Simultaneously, this therapy activates immune responses and mobilizes immune cells, thereby eliminating residual or distant cancer cells. However, because of OVs' high immunogenicity and immune clearance during circulation, their clinical applications are currently limited to intratumoral injections, and their use is severely restricted. In recent years, numerous studies have used nanomaterials to modify OVs to decrease virulence and increase safety for intravenous injection. The most commonly used nanomaterials for modifying OVs are liposomes, polymers, and albumin, because of their biosafety, practicability, and effectiveness. The aim of this review is to summarize progress in the use of these nanomaterials in preclinical experiments to modify OVs and to discuss the challenges encountered from basic research to clinical application.
Collapse
Affiliation(s)
- Liting Chen
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Phase I Clinical Trials Center, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang 110102, China
- 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
| | - Zhijun Ma
- Department of General Surgery, Panjin People’s Hospital, Panjin 124221, China
| | - Chen Xu
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Phase I Clinical Trials Center, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang 110102, China
- 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
| | - Youbang Xie
- Department of Hematology and Rheumatology, Qinghai Provincial People’s Hospital, Xining 810007, China
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau 999078, China
| | - Shuhui Song
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Phase I Clinical Trials Center, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang 110102, China
| | - Xiao Zhao
- 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
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Phase I Clinical Trials Center, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang 110102, China
| |
Collapse
|
4
|
Lee J, Kwon YE, Edwards SD, Guim H, Jae Jeong K. Improved biocompatibility of dendrimer-based gene delivery by histidine-modified nuclear localization signals. Int J Pharm 2023; 644:123299. [PMID: 37558147 DOI: 10.1016/j.ijpharm.2023.123299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/29/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023]
Abstract
Polyamidoamine (PAMAM) dendrimers have been explored as an alternative to polyethylenimine (PEI) as a gene delivery carrier because of their relatively low cytotoxicity and excellent biocompatibility. The transfection efficiency of PAMAM dendrimers can be improved by the addition of nuclear localization signal (NLS), a positively charged peptide sequence recognized by cargo proteins in the cytoplasm for nuclear transport. However, increased positive charges from NLS can cause damage to the cytoplasmic and mitochondrial membranes and lead to reactive oxygen species (ROS)-induced cytotoxicity. This negative effect of NLS can be negated without a significant reduction in transfection efficiency by adding histidine, an essential amino acid known as a natural antioxidant, to NLS. However, little is known about the exact mechanism by which histidine reduces cytotoxicity of NLS-modified dendrimers. In this study, we selected cystamine core PAMAM dendrimer generation 2 (cPG2) and conjugated it with NLS derived from Merkel cell polyomavirus large T antigen and histidine (n = 0-3) to improve transfection efficiency and reduce cytoxicity. NLS-modified cPG2 derivatives showed similar or higher transfection efficiency than PEI 25 kDa in NIH3T3 and human mesenchymal stem cells (hMSC). The cytotoxicity of NLS-modified cPG2 derivatives was substantially lower than PEI 25 kDa and was further reduced as the number of histidine in NLS increased. To understand the mechanism of cytoprotective effect of histidine-conjugated NLS, we examined ROS scavenging, hydroxyl radical generation and mitochondrial membrane potential as a function of the number of histidine in NLS. As the number of hisidine increased, cPG2 scavenged ROS more effectively as evidenced by the hydroxyl radical antioxidant capacity (HORAC) assay. This was consistent with the reduced intracellular hydroxyl radical concentration measured by 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) assay in NIH3T3. Finally, fluorescence imaging with JC-1 confirmed that the mitochondrial membranes of NIH 3T3 were well-protected during the transfection when NLS contained histidine. These experimental results confirm the hypothesis that histidine residues scavenge ROS that is generated during the transfection process, preventing the excessive damage to mitochondrial membranes, leading to reduced cytotoxicity.
Collapse
Affiliation(s)
- Jeil Lee
- Department of Chemical Engineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| | - Yong-Eun Kwon
- Center for Scientific Instrumentation, Korea Basic Science Institute, 169-148 Gwahak-ro, Yuseong-gu, Daejeon 34133, Republic of Korea
| | - Seth D Edwards
- Department of Chemical Engineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| | - Hwanuk Guim
- Research Center for Materials Analysis, Korea Basic Science Institute, 169-148 Gwahak-ro, Yuseong-gu, Daejeon 34133, Republic of Korea
| | - Kyung Jae Jeong
- Department of Chemical Engineering, University of New Hampshire, Durham, New Hampshire 03824, United States.
| |
Collapse
|
5
|
Wang Y, Wang C, Sylvers J, Segura T, Yuan F. Nanoenhancer for improving naked DNA electrotransfection In vivo. Front Bioeng Biotechnol 2023; 11:1181795. [PMID: 37229498 PMCID: PMC10203387 DOI: 10.3389/fbioe.2023.1181795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction: Electrotransfection (ET) is a non-viral approach widely used for delivery of naked nucleic acids. Its efficiency can be increased in vitro by treatment of cells with various small molecule enhancers. However, these enhancers often fail to improve ET in vivo, presumably due to rapid clearance in tissues after local injection, reducing their cellular uptake. To this end, we propose to develop a new type of ET enhancers, which we term nanoenhancer, that diffuse slowly in tissues and are poorly absorbed by blood and lymph microvessels. Methods: Two nanoenhancers were synthesized with alginate (Alg) and chitosan (Chi) with or without poly (ethylene imine) (PEI). They were used to treat cells in vitro or mouse muscle in the hind leg in vivo prior to ET of plasmid DNA coding reporter genes. At 24 hours post ET, the efficiency of ET was quantified, and compared with that in the untreated controls. Changes in lysosomal size and acidity post nanoenhancer treatment were measured with fluorescence microscopy techniques. Results and discussion: We observed that the pretreatment of cells with the nanoenhancers could enhance the ET efficiency and cell viability in both C2C12 and HCT116 cells in vitro, and the nanoenhancer pretreatment had similar effects on the ET efficiency in vivo. Mechanisms of the enhancement were related to transient inactivation of lysosomal functions triggered by the nanoenhancer treatment. The concept of nanoenhancer will lead to development of new enhancers that can be used to improve ET efficiency in vivo, highlighting its potential in clinical applications.
Collapse
|
6
|
Polyethyleneimine-Based Drug Delivery Systems for Cancer Theranostics. J Funct Biomater 2022; 14:jfb14010012. [PMID: 36662059 PMCID: PMC9862060 DOI: 10.3390/jfb14010012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
With the development of nanotechnology, various types of polymer-based drug delivery systems have been designed for biomedical applications. Polymer-based drug delivery systems with desirable biocompatibility can be efficiently delivered to tumor sites with passive or targeted effects and combined with other therapeutic and imaging agents for cancer theranostics. As an effective vehicle for drug and gene delivery, polyethyleneimine (PEI) has been extensively studied due to its rich surface amines and excellent water solubility. In this work, we summarize the surface modifications of PEI to enhance biocompatibility and functionalization. Additionally, the synthesis of PEI-based nanoparticles is discussed. We further review the applications of PEI-based drug delivery systems in cancer treatment, cancer imaging, and cancer theranostics. Finally, we thoroughly consider the outlook and challenges relating to PEI-based drug delivery systems.
Collapse
|
7
|
Nguyen Le NM, Zsák S, Le-Vinh B, Friedl JD, Kali G, Knoll P, Seitter HW, Koschak A, Bernkop-Schnürch A. Charge-Converting Nanoemulsions as Promising Retinal Drug and Gene Delivery Systems. ACS APPLIED MATERIALS & INTERFACES 2022; 14:44981-44991. [PMID: 36125912 PMCID: PMC9542710 DOI: 10.1021/acsami.2c11649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/08/2022] [Indexed: 05/31/2023]
Abstract
AIM This study aimed to develop phosphatase-responsive ζ potential converting nanocarriers utilizing polyphosphate-coated cell-penetrating peptide (CPP)-decorated nanoemulsions (NEs) as a novel gene delivery system to retinal cells. METHODS Poly-l-lysine (PLL) was first conjugated with oleylamine (OA) only at its carboxylic end to form the amphiphilic PLL-oleylamine (PLOA) conjugate. Afterward, NEs were loaded with PLOA prior to being coated with tripolyphosphate (TPP) to generate PLOA/TPP NEs. A plasmid containing a reporter gene for green fluorescent protein plasmid (pGFP) was complexed with cationic surfactants forming hydrophobic ion pairs that were loaded in the oily core of NEs. Phosphate removal, ζ potential conversion, and cytotoxicity of the system were evaluated. Cellular uptake and transfection efficiency were investigated in 661W photoreceptor-like cells via microscopic analysis, fluorescence spectroscopy, and flow cytometry. RESULTS Dephosphorylation of PLOA/TPP NEs triggered by alkaline phosphatase (ALP) resulted in the exposure of positive amine groups on the surface of NE droplets and a notable conversion of the ζ potential from -22.4 to +8.5 mV. Cellular uptake of PLOA/TPP NEs performed on 661W photoreceptor-like cells showed a 3-fold increase compared to control NEs. Furthermore, PLOA/TPP NEs also showed low cytotoxicity and high transfection efficacy with ∼50% of cells transfected. CONCLUSIONS Polyphosphate-coated CPP-decorated NEs triggered by ALP could be a promising nanosystem to efficiently deliver drugs and genetic materials to photoreceptor-like cells and other retinal cells for potential treatments of retinal diseases.
Collapse
Affiliation(s)
- Nguyet-Minh Nguyen Le
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Sarah Zsák
- Center
for Chemistry and Biomedicine, Department of Pharmacology and Toxicology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Bao Le-Vinh
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Julian David Friedl
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Gergely Kali
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Patrick Knoll
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Hartwig Wolfram Seitter
- Center
for Chemistry and Biomedicine, Department of Pharmacology and Toxicology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Alexandra Koschak
- Center
for Chemistry and Biomedicine, Department of Pharmacology and Toxicology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| |
Collapse
|
8
|
Thambi T, Hong J, Yoon AR, Yun CO. Challenges and progress toward tumor-targeted therapy by systemic delivery of polymer-complexed oncolytic adenoviruses. Cancer Gene Ther 2022; 29:1321-1331. [PMID: 35444290 PMCID: PMC9576595 DOI: 10.1038/s41417-022-00469-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/10/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022]
Abstract
Oncolytic adenovirus (oAd) elicits antitumor activity by preferential viral replication in cancer cells. However, poor systemic administrability or suboptimal intratumoral retainment of the virus remains a major challenge toward maximizing the antitumor activity of oAd in a clinical environment. To surmount these issues, a variety of non-immunogenic polymers has been used to modify the surface of oAds chemically or physically. Complexation of oAd with polymers can effectively evade the host immune response and reduces nonspecific liver sequestration. The tumor-specific delivery of these complexes can be further improved upon by inclusion of tumor-targeting moieties on the surface. Therefore, modification of the Ad surface using polymers is viewed as a potential strategy to enhance the delivery of Ad via systemic administration. This review aims to provide a comprehensive overview of polymer-complexed Ads, their progress, and future challenges in cancer treatment.
Collapse
Affiliation(s)
- Thavasyappan Thambi
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro Seongdong-gu, Seoul, 04763, Korea
| | - JinWoo Hong
- GeneMedicine CO., Ltd., 222 Wangsimni-ro Seongdong-gu, Seoul, 04763, Seoul, Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro Seongdong-gu, Seoul, 04763, Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, 04763, Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro Seongdong-gu, Seoul, 04763, Korea.
- GeneMedicine CO., Ltd., 222 Wangsimni-ro Seongdong-gu, Seoul, 04763, Seoul, Korea.
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, 04763, Korea.
- Institute of Nano Science and Technology (INST), Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Korea.
| |
Collapse
|
9
|
Shams A, Shabani R, Asgari H, Karimi M, Najafi M, Asghari-Jafarabadi M, Razavi SM, Miri SR, Abbasi M, Mohammadi A, Koruji M. In vitro elimination of EL4 cancer cells from spermatogonia stem cells by miRNA-143- and 206-loaded folic acid conjugated PLGA nanoparticles. Nanomedicine (Lond) 2022; 17:531-545. [PMID: 35264013 DOI: 10.2217/nnm-2021-0210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: MiRNA's-143 and -206 are powerful apoptotic regulators in cancer cells. This study aimed to use miRNA-143- and 206-loaded poly(lactic-co-glycolic) acid (PLGA) nanoparticles conjugated with folic acid to induce apoptosis in the EL4 cancer cells. Materials & methods: The therapy was conducted in six groups: Treatment with both miRNAs simultaneously (mixed miRNAs), miRNA-206 treatment, miRNA-143 treatment, blank PLGA, blank polyethylenimine (PEI) and complex PEI-miRNAs. Results: In terms of viability, in mixed miRNAs, no synergistic effect was observed on EL4 cell elimination. However, in the single miRNA-206 group, a stronger apoptotic effect was observed than the mixed miRNAs group and single miRNA-143 group alone. Conclusion: MiRNAs' apoptotic induction effects in cancer cells were found to be remarkable.
Collapse
Affiliation(s)
- Azar Shams
- Stem cell & Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ronak Shabani
- Stem cell & Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Asgari
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdi Karimi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Asghari-Jafarabadi
- Department of Statistics & Epidemiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Road Traffic Injury Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Mohsen Razavi
- Clinic of Hematology & Oncology, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Rouhollah Miri
- Department of Surgical Oncology, Cancer Institute,Tehran University of Medical Science, Tehran, Iran
| | - Mehdi Abbasi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Mohammadi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Koruji
- Stem cell & Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Huang Z, Xiao YP, Guo Y, Yang HZ, Zhao RM, Zhang J, Yu XQ. A cyclen-based fluoropolymer as a versatile vector for gene and protein delivery. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
11
|
Hooshmand SE, Sabet MJ, Hasanzadeh A, Mousavi SMK, Moghadam NH, Hooshmand SA, Rabiee N, Liu Y, Hamblin MR, Karimi M. Histidine‐enhanced gene delivery systems: The state of the art. J Gene Med 2022; 24:e3415. [DOI: 10.1002/jgm.3415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/26/2022] [Accepted: 01/29/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Seyyed Emad Hooshmand
- 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
| | - Makkieh Jahanpeimay Sabet
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Akbar Hasanzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Seyede Mahtab Kamrani Mousavi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Niloofar Haeri Moghadam
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Seyed Aghil Hooshmand
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics University of Tehran Tehran Iran
| | - Navid Rabiee
- Department of Physics Sharif University of Technology Tehran Iran
- School of Engineering Macquarie University Sydney New South Wales Australia
| | - Yong Liu
- Institute of Functional Nano & Soft Materials (FUNSOM) Soochow University Suzhou Jiangsu China
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science University of Johannesburg South Africa
| | - Mahdi Karimi
- 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
- Oncopathology Research Center Iran University of Medical Sciences Tehran Iran
- Research Center for Science and Technology in Medicine Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
12
|
Wang P, Yang P, Qian K, Li Y, Xu S, Meng R, Guo Q, Cheng Y, Cao J, Xu M, Lu W, Zhang Q. Precise gene delivery systems with detachable albumin shell remodeling dysfunctional microglia by TREM2 for treatment of Alzheimer's disease. Biomaterials 2021; 281:121360. [PMID: 34991033 DOI: 10.1016/j.biomaterials.2021.121360] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 12/19/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022]
Abstract
Intervention of the over-activated microglia-aggravated neuroinflammation represents a promising therapeutic strategy for Alzheimer's disease (AD). Upregulation of triggering receptor expressed on myeloid cells-2 (TREM2) attenuates the neuroinflammatory processes and normalizes the dysfunctional microglia. However, Trem2-gene therapy for AD by the effective non-invasive delivery systems is unexploited. Herein, we report the microglia-targeted gene delivery systems (PHSA@PF/pTREM2) composed of a core of fluorinated polyethylenimine condensing the TREM2-encoding plasmid (PF/pTREM2) and a shell of human serum albumin conjugated with both cis-aconitic anhydride and neural cell adhesion molecule-mimetic peptide P2 (PHSA). Thanks to the shedding effect of the albumin coated, PHSA@PF/pTREM2 exhibit prolonged blood circulation and low cytotoxicity. PHSA@PF/pTREM2 achieve brain accumulation as high as 2.17% injected dose per gram of brain and the microglial-targeting effect (targeting specificity of 41.9%) via the systemic administration. The nanocomplexes can be detached PHSA-shell in the acidic endo-lysosomes via the cleavage of cis-aconitic amide bond, resulting in PF/pTREM2 exposure for efficient endo-lysosomal escape and gene transfection. PHSA@PF/pTREM2 upregulate the TREM2 level and regulate microglial polarization toward M2-phenotype for remodeling the inflammatory microenvironment and enhanced Aβ clearance, leading to an improvement of cognitive performance in APP/PS1 mice. This work provides a promising gene delivery platform to reverse dysfunctional microglia for AD therapy.
Collapse
Affiliation(s)
- Pengzhen Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Shuting Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Qian Guo
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Jinxu Cao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Wei Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China.
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, PR China.
| |
Collapse
|
13
|
Diaz IL, Jérôme V, Freitag R, Perez LD. Development of poly(ethyleneimine) grafted amphiphilic copolymers: Evaluation of their cytotoxicity and ability to complex DNA. J BIOACT COMPAT POL 2021. [DOI: 10.1177/08839115211053925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Poly(ethyleneimine) (PEI) is one of the most widely used cationic polymers for gene delivery. The high molecular weight polymer, which is commercially available, is highly efficient but also very cytotoxic. The reduction in charge density by using nonlinear architectures based on low molecular weight (LMW) PEI is a promising approach to produce safer DNA-vectors. Herein, a group of cationic graft copolymers with different composition containing a hydrophobic biocompatible backbone and LMW linear PEI (lPEI) grafts obtained by ring opening polymerization and click chemistry was studied. The self-assembly and DNA complexation behavior of these materials was analyzed by the gel retardation assay, zeta potential measurements, and dynamic light scattering. The copolymers formed positively charged particles in water with average sizes between 270 and 377 nm. After they were added to DNA in serum-free medium, these particles acquired negative/near-neutral charges and increased in size depending on the N/P ratio. All copolymers showed reduced cytotoxicity compared to the 25 kDa lPEI used as reference, but the transfection efficiency was reduced. This result suggested that the cationic segments were too small to fully condense the DNA and promote cellular uptake, even with the use of several grafts and the introduction of hydrophobic domains. The trends found in this research showed that a higher degree of hydrophobicity and a higher grafting density can enhance the interaction between the copolymers and DNA. These trends could direct further structural modifications in the search for effective and safe vectors based on this polycation.
Collapse
Affiliation(s)
- Ivonne L Diaz
- Departamento de Química, Universidad Nacional de Colombia, Bogotá DC, Colombia
| | - Valérie Jérôme
- Process Biotechnology, University of Bayreuth, Bayreuth, Germany
| | - Ruth Freitag
- Process Biotechnology, University of Bayreuth, Bayreuth, Germany
| | - León D Perez
- Departamento de Química, Universidad Nacional de Colombia, Bogotá DC, Colombia
| |
Collapse
|
14
|
Liufu C, Li Y, Lin Y, Yu J, Du M, Chen Y, Yang Y, Gong X, Chen Z. Synergistic ultrasonic biophysical effect-responsive nanoparticles for enhanced gene delivery to ovarian cancer stem cells. Drug Deliv 2021; 27:1018-1033. [PMID: 32627597 PMCID: PMC8216435 DOI: 10.1080/10717544.2020.1785583] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer stem cells (OCSCs) that are a subpopulation within bulk tumor survive chemotherapy and conduce to chemo-resistance and tumor relapse. However, conventional gene delivery is unsuitable for the on-demand content release, which limits OCSCs therapeutic utility. Here, we reported ultrasound-targeted microbubble destruction (UTMD)-triggerable poly(ethylene glycol)-disulfide bond-polyethylenimine loaded microbubble (PSP@MB). Taking advantage of glutathione (GSH) responsiveness, ultrasound triggering and spatiotemporally controlled release manner, PSP@MB is expected to realize local gene delivery for OCSCs treatment. But the biophysical mechanisms of gene delivery via PSP@MB and ultrasound remain unknown. The aim of this study is to determine the potential of gene delivery to OCSCs via ultrasonic synergistic biophysical effects and GSH-sensitive PSP@MB. The GSH-sensitive disulfide bond cleavable properties of PSP@MB were confirmed by 1H NMR spectra and infrared spectroscopy. The biophysical mechanisms between PSP@MB and cells were confirmed by scanning electron microscopy (SEM) and confocal laser scanning microscope (CLSM) to optimize the ultrasonic gene delivery system. The gene transfection via ultrasound and PSP@MB was closely related to the biophysical mechanisms (sonoporation, enhanced-endocytosis, sonoprinting, and endosomal escape). Ultrasound combined with PSP@MB successfully delivered aldehyde dehydrogenase 1 (ALDH1) short hairpin RNA (shRNA) plasmid to OCSCs and promoted apoptosis of OCSCs. The gene transfection rate and apoptosis rate were (18.41 ± 2.41)% and (32.62 ± 2.36)% analyzed by flow cytometry separately. This study showed that ultrasound triggering and GSH responsive PSP@MB might provide a novel strategy for OCSCs treatment via sonoporation and enhanced-endocytosis.
Collapse
Affiliation(s)
- Chun Liufu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yue Li
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yan Lin
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinsui Yu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meng Du
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuhao Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yaozhang Yang
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaojing Gong
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen University Town, Shenzhen, China
| | - Zhiyi Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
15
|
A novel UTMD system facilitating nucleic acid delivery into MDA-MB-231 cells. Biosci Rep 2021; 40:221955. [PMID: 31990029 PMCID: PMC7029150 DOI: 10.1042/bsr20192573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/26/2019] [Accepted: 01/13/2020] [Indexed: 01/13/2023] Open
Abstract
Gene therapy is emerging as a promising method for the treatment of various diseases. The safe and efficient delivery of therapeutic nucleic acids is a gene therapy prerequisite. Ultrasound, particularly in combination with microbubbles composed of biocompatible materials such as lipid, PLGA and chitosan, is a novel non-viral tool for gene transportation. Under ultrasound irradiation, microbubbles explode and generate pores in the cell membrane. Hence, genes can enter cells more easily. In order to transfect nucleic acids into MDA-MB-231 cells in a low-cost and non-viral manner for further breast cancer gene therapy studies, we explored ultrasound targeted microbubble destruction (UTMD) technology and evaluated the efficiency and safety of the delivery of plasmid encoding enhanced green fluorescent protein (pEGFP) and a microRNA-34a (miR-34a) mimic by UTMD. Sonovitro ultrasonic apparatus was employed to generate ultrasonic field, which was developed by our group. Ultrasonic parameters, including acoustic intensity (AI), exposure time (ET) and duty cycle (DC), were optimized at 0.6 W/cm2 AI, 20 s ET and 20% DC, the cell viability was not obviously impaired. Under these conditions, the UTMD-mediated transfection efficiency of pEGFP was greater than 40%. In addition to plasmid DNA, an miR-34a mimic was also successfully introduced into the cytoplasm by UTMD and found to inhibit proliferation, induce apoptosis of MDA-MB-231 cells and regulate downstream molecules. The present study indicates that further in vivo UTMD-mediated gene therapy studies are warranted.
Collapse
|
16
|
Ullah I, Zhao J, Su B, Rukh S, Guo J, Ren XK, Xia S, Zhang W, Feng Y. Redox stimulus disulfide conjugated polyethyleneimine as a shuttle for gene transfer. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:118. [PMID: 33247778 DOI: 10.1007/s10856-020-06457-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 10/27/2020] [Indexed: 06/12/2023]
Abstract
Redox-responsive cationic polymers have gained considerable attention in gene delivery due to low cytotoxicity and spatio-temporal release of DNA into the cells. Here, we reported the synthesis of reducible disulfide conjugated polyethyleneimine (1.8 kDa) (denoted as SS-PEI) and its application to transfer pEGFP-ZNF580 plasmid (pZNF580) into EA.hy926 cell. This reducible SS-PEI polymer was prepared by one-step polycondensation reaction of low molecular weight PEI with bis-(p-nitrophenyl)-3,3'-dithiodipropionate. The SS-PEI successfully condensed pZNF580 into nano-sized complexes (170 ± 1.5 nm to 255 ± 1.6 nm) with zeta potentials of 3 ± 0.4 mV to 17 ± 0.9 mV. The complexes could be triggered to release pZNF580 when exposed to the reducing environment of 5 mM dithiothreitol. Besides, the SS-PEI exhibited low cytotoxicity. In vitro transfection results showed that SS-PEI exhibited good transfection efficiency comparable to PEI25kDa. Thus, the SS-PEI could act as an reducible gene carrier with good transfection efficiency and low cytotoxicity.
Collapse
Affiliation(s)
- Ihsan Ullah
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, China
| | - Jing Zhao
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, China
| | - Bin Su
- Department of Clinical Research, Characteristic Medical Center of Chinese People's Armed Police Force, 220 Chenglin Road, Tianjin, 300162, China
| | - Shah Rukh
- Department of Chemistry, School of Science, Abdul Wali Khan University, Mardan, 23200, Pakistan
| | - Jintang Guo
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, China
- Collaborative Innovation Centre of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin, 300072, China
| | - Xiang-Kui Ren
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, China
- Collaborative Innovation Centre of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin, 300072, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Weijin Road 92, Tianjin, 300072, China
| | - Shihai Xia
- Department of Hepatopancreatobiliary and Splenic Medicine, Affiliated Hospital, Logistics University of People's Armed Police Force, Chenglin Road 220, Tianjin, 300162, China
| | - Wencheng Zhang
- Department of Physiology and Pathophysiology, Logistics University of People's Armed Police Force, Tianjin, 300309, China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, China.
- Department of Clinical Research, Characteristic Medical Center of Chinese People's Armed Police Force, 220 Chenglin Road, Tianjin, 300162, China.
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Weijin Road 92, Tianjin, 300072, China.
| |
Collapse
|
17
|
Niu G, Jin Z, Zhang C, He D, Gao X, Zou C, Zhang W, Ding J, Das BC, Severinov K, Hitzeroth II, Debata PR, Ma X, Tian X, Gao Q, Wu J, You Z, Tian R, Cui Z, Fan W, Xie W, Huang Z, Cao C, Xu W, Xie H, Xu H, Tang X, Wang Y, Yu Z, Han H, Tan S, Chen S, Hu Z. An effective vaginal gel to deliver CRISPR/Cas9 system encapsulated in poly (β-amino ester) nanoparticles for vaginal gene therapy. EBioMedicine 2020; 58:102897. [PMID: 32711250 PMCID: PMC7387785 DOI: 10.1016/j.ebiom.2020.102897] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Gene therapy has held promises for treating specific genetic diseases. However, the key to clinical application depends on effective gene delivery. METHODS Using a large animal model, we developed two pharmaceutical formulations for gene delivery in the pigs' vagina, which were made up of poly (β-amino ester) (PBAE)-plasmid polyplex nanoparticles (NPs) based two gel materials, modified montmorillonite (mMMT) and hectorite (HTT). FINDINGS By conducting flow cytometry of the cervical cells, we found that PBAE-GFP-NPs-mMMT gel was more efficient than PBAE-GFP-NPs-HTT gel in delivering exogenous DNA intravaginally. Next, we designed specific CRISPR/SpCas9 sgRNAs targeting porcine endogenous retroviruses (PERVs) and evaluated the genome editing efficacy in vivo. We discovered that PERV copy number in vaginal epithelium could be significantly reduced by the local delivery of the PBAE-SpCas9/sgRNA NPs-mMMT gel. Comparable genome editing results were also obtained by high-fidelity version of SpCas9, SpCas9-HF1 and eSpCas9, in the mMMT gel. Further, we confirmed that the expression of topically delivered SpCas9 was limited to the vagina/cervix and did not diffuse to nearby organs, which was relatively safe with low toxicity. INTERPRETATION Our data suggested that the PBAE-NPs mMMT vaginal gel is an effective preparation for local gene therapy, yielding insights into novel therapeutic approaches to sexually transmitted disease in the genital tract. FUNDING This work was supported by the National Science and Technology Major Project of the Ministry of science and technology of China (No. 2018ZX10301402); the National Natural Science Foundation of China (81761148025, 81871473 and 81402158); Guangzhou Science and Technology Programme (No. 201704020093); National Ten Thousand Plan-Young Top Talents of China, Fundamental Research Funds for the Central Universities (17ykzd15 and 19ykyjs07); Three Big Constructions-Supercomputing Application Cultivation Projects sponsored by National Supercomputer Center In Guangzhou; the National Research FFoundation (NRF) South Africa under BRICS Multilateral Joint Call for Proposals; grant 17-54-80078 from the Russian Foundation for Basic Research.
Collapse
Affiliation(s)
- Gang Niu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhuang Jin
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Chong Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dan He
- Department of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Xueqin Gao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chenming Zou
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiahui Ding
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bhudev C Das
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Uttar Pradesh, Noida 201313, India
| | - Konstantin Severinov
- Skolkovo Institute of Science and Technology, Skolkovo, Moscow Region 143025, Russian Federation
| | - Inga Isabel Hitzeroth
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Cape Town 7701, South Africa
| | - Priya Ranjan Debata
- Department of Zoology, North Orissa University, Takatpur, Baripada, Odisha 757003, India
| | - Xin Ma
- Department of Urology, General Hospital of People's Liberation Army, Beijing 100039, China
| | - Xun Tian
- Department of Obstetrics and Gynecology, Academician expert workstation, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei, China
| | - Qinglei Gao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jun Wu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, Guangdong, China
| | - Zeshan You
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Rui Tian
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zifeng Cui
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Weiwen Fan
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Weiling Xie
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhaoyue Huang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Chen Cao
- Department of Obstetrics and Gynecology, Academician expert workstation, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei, China
| | - Wei Xu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Hongxian Xie
- Generulor Company Bio-X Lab, Guangzhou 510006, Guangdong, China
| | - Hongyan Xu
- Department of Obstetrics and Gynecology, Yuebei People's Hospital, Medical College of Shantou University, Shaoguan 512026, Guangdong, China
| | - Xiongzhi Tang
- Department of Obstetrics and Gynecology, Guilin People's Hospital, Guilin, The Guangxi Zhuang Autonomous Region, 541002, China
| | - Yan Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Zhiying Yu
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Shenzhen University, Shenzhen 518000, Guangdong, China
| | - Hui Han
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine & Department of Urology, Yat-sen University Cancer Center, Guangzhou 510080, Guangdong Province, China
| | - Songwei Tan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Shuqin Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| | - Zheng Hu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; Precision Medicine Institute, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
18
|
Cao M, Gao Y, Qiu N, Shen Y, Shen P. Folic acid directly modified low molecular weight of polyethyleneimine for targeted pDNA delivery. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
19
|
Jahanafrooz Z, Baradaran B, Mosafer J, Hashemzaei M, Rezaei T, Mokhtarzadeh A, Hamblin MR. Comparison of DNA and mRNA vaccines against cancer. Drug Discov Today 2020; 25:552-560. [PMID: 31843577 PMCID: PMC7080609 DOI: 10.1016/j.drudis.2019.12.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/23/2019] [Accepted: 12/06/2019] [Indexed: 12/15/2022]
Abstract
Nucleic acid vaccines (NAVs) have recently been tested as a cancer therapy. DNA and mRNA vaccines deliver genetic information encoding tumor antigens (TAs) to the host, which then produces immune responses against cancer cells that express the TAs. Although NAVs are easy, safe, and simple to manufacture, they have not so far been considered viable alternatives to peptide vaccines. Choosing the right TAs, insufficient immunogenicity, and the immunosuppressive nature of cancer are some challenges to this approach. In this review, we discuss approaches that been used to improve the efficiency of anticancer NAVs.
Collapse
Affiliation(s)
- Zohreh Jahanafrooz
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Mosafer
- Department of Medical Biotechnology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mahmoud Hashemzaei
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Tayebeh Rezaei
- Department of Molecular Medicine and Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| |
Collapse
|
20
|
Non-Viral in Vitro Gene Delivery: It is Now Time to Set the Bar! Pharmaceutics 2020; 12:pharmaceutics12020183. [PMID: 32098191 PMCID: PMC7076396 DOI: 10.3390/pharmaceutics12020183] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 01/31/2023] Open
Abstract
Transfection by means of non-viral gene delivery vectors is the cornerstone of modern gene delivery. Despite the resources poured into the development of ever more effective transfectants, improvement is still slow and limited. Of note, the performance of any gene delivery vector in vitro is strictly dependent on several experimental conditions specific to each laboratory. The lack of standard tests has thus largely contributed to the flood of inconsistent data underpinning the reproducibility crisis. A way researchers seek to address this issue is by gauging the effectiveness of newly synthesized gene delivery vectors with respect to benchmarks of seemingly well-known behavior. However, the performance of such reference molecules is also affected by the testing conditions. This survey points to non-standardized transfection settings and limited information on variables deemed relevant in this context as the major cause of such misalignments. This review provides a catalog of conditions optimized for the gold standard and internal reference, 25 kDa polyethyleneimine, that can be profitably replicated across studies for the sake of comparison. Overall, we wish to pave the way for the implementation of standardized protocols in order to make the evaluation of the effectiveness of transfectants as unbiased as possible.
Collapse
|
21
|
Cheng YC, Guo SL, Chung KD, Hu WW. Electrical Field-Assisted Gene Delivery from Polyelectrolyte Multilayers. Polymers (Basel) 2020; 12:E133. [PMID: 31935814 PMCID: PMC7022892 DOI: 10.3390/polym12010133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/19/2019] [Accepted: 01/01/2020] [Indexed: 12/12/2022] Open
Abstract
To sustain gene delivery and elongate transgene expression, plasmid DNA and cationic nonviral vectors can be deposited through layer-by-layer (LbL) assembly to form polyelectrolyte multilayers (PEMs). Although these macromolecules can be released for transfection purposes, their entanglement only allows partial delivery. Therefore, how to efficiently deliver immobilized genes from PEMs remains a challenge. In this study, we attempt to facilitate their delivery through the pretreatment of the external electrical field. Multilayers of polyethylenimine (PEI) and DNA were deposited onto conductive polypyrrole (PPy), which were placed in an aqueous environment to examine their release after electric field pretreatment. Only the electric field perpendicular to the substrate with constant voltage efficiently promoted the release of PEI and DNA from PEMs, and the higher potential resulted in the more releases which were enhanced with treatment time. The roughness of PEMs also increased after electric field treatment because the electrical field not only caused electrophoresis of polyelectrolytes and but also allowed electrochemical reaction on the PPy electrode. Finally, the released DNA and PEI were used for transfection. Polyplexes were successfully formed after electric field treatment, and the transfection efficiency was also improved, suggesting that this electric field pretreatment effectively assists gene delivery from PEMs and should be beneficial to regenerative medicine application.
Collapse
Affiliation(s)
- Yu-Che Cheng
- Proteomics Laboratory, Department of Medical Research, Cathay General Hospital, Taipei 10630, Taiwan;
- Department of Biomedical Sciences and Engineering, National Central University, Zhongli District, Taoyuan City 32001, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
| | - Shu-Lin Guo
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
- Department of Anesthesiology, Cathay General Hospital, Taipei 10630, Taiwan
- Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei 11490, Taiwan
| | - Kun-Da Chung
- Department of Chemical and Materials Engineering, National Central University, Zhongli District, Taoyuan City 32001, Taiwan
| | - Wei-Wen Hu
- Department of Chemical and Materials Engineering, National Central University, Zhongli District, Taoyuan City 32001, Taiwan
| |
Collapse
|
22
|
Liufu C, Li Y, Tu J, Zhang H, Yu J, Wang Y, Huang P, Chen Z. Echogenic PEGylated PEI-Loaded Microbubble As Efficient Gene Delivery System. Int J Nanomedicine 2019; 14:8923-8941. [PMID: 31814720 PMCID: PMC6863126 DOI: 10.2147/ijn.s217338] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 09/02/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are responsible for cancer therapeutic resistance and metastasis. To date, in addition to surgery, chemotherapy, and radiotherapy, gene delivery has emerged as a potential therapeutic modality for ovarian cancer. Efficient and safe targeted gene delivery is complicated due to the tumor heterogeneity barrier. Ultrasound (US)-stimulated microbubbles (MBs) have demonstrated a method of enabling non-invasive targeted gene delivery. PURPOSE The purpose of our study was to show the utility of poly(ethylene glycol)-SS-polyethylenimine-loaded microbubbles (PSP@MB) as an ultrasound theranostic and redox-responsive agent in a gene delivery system. PATIENTS AND METHODS PSP nanoparticles were conjugated to the MB surface through biotin-avidin linkage, increasing the gene-loading efficiency of MB. The significant increase in the release of genes from the PSP@MB complexes was achieved upon ultrasound exposure. The positive surface charge in PSP@MB can condense the plasmid through electrostatic interactions; agarose-gel electrophoresis further confirmed the ability of PSP@MB to condense plasmids. The morphology, particle sizes and zeta potential of PSP@MB were characterized by transmission electron microscopy and dynamic light scattering. RESULTS Laser confocal microscopy showed that the combination of ultrasound with PSP@MB could promote the cellular uptake of plasmids. Plasmids which encode enhanced green fluorescence protein (EGFP) reporter genes or luciferase reporter genes were delivered to CSCs in vitro and to subcutaneous xenografts in vivo via the combination of ultrasound with PSP@MB. Gene transfection efficiency was evaluated by fluorescence microscopy and In Vivo Imaging Systems. This study demonstrated that the combination of ultrasound with PSP@MB can remarkably promote gene delivery to solid tumors as well as diminishing the toxicity towards normal tissues in vivo. The combination of PSP@MB and the use of ultrasound can efficiently enhance accumulation, extravasation and penetration into solid tumors. CONCLUSION Taken together, our study showed that this novel PSP@MB and ultrasound-mediated gene delivery system could efficiently target CSCs.
Collapse
Affiliation(s)
- Chun Liufu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Experimental Center, The Liwan Hospital of The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong510000, People’s Republic of China
| | - Yue Li
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Experimental Center, The Liwan Hospital of The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong510000, People’s Republic of China
| | - Jiawei Tu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Experimental Center, The Liwan Hospital of The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong510000, People’s Republic of China
| | - Hui Zhang
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Experimental Center, The Liwan Hospital of The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong510000, People’s Republic of China
| | - Jinsui Yu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Experimental Center, The Liwan Hospital of The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong510000, People’s Republic of China
| | - Yi Wang
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Experimental Center, The Liwan Hospital of The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong510000, People’s Republic of China
| | - Pintong Huang
- Department of Ultrasound, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Zhiyi Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Experimental Center, The Liwan Hospital of The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong510000, People’s Republic of China
| |
Collapse
|
23
|
Zhang H, Rao F, Chen Z, Wang Y, Li Y. MicroRNA-34a and E4orf4 synergistically promote apoptosis in Hela cells. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1673206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Hui Zhang
- Laboratory of Ultrasound Molecular Imaging, Department of Ultrasound Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
- Laboratory of Ultrasound Molecular Imaging, Department of Ultrasound Medicine, The Liwan Hospital, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Fang Rao
- Laboratory of Ultrasound Molecular Imaging, Department of Ultrasound Medicine, The Liwan Hospital, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Zhiyi Chen
- Laboratory of Ultrasound Molecular Imaging, Department of Ultrasound Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
- Laboratory of Ultrasound Molecular Imaging, Department of Ultrasound Medicine, The Liwan Hospital, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Yi Wang
- Laboratory of Ultrasound Molecular Imaging, Department of Ultrasound Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Yue Li
- Laboratory of Ultrasound Molecular Imaging, Department of Ultrasound Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| |
Collapse
|