1
|
Mohammadi N, Fayazi Hosseini N, Nemati H, Moradi-Sardareh H, Nabi-Afjadi M, Kardar GA. Revisiting of Properties and Modified Polyethylenimine-Based Cancer Gene Delivery Systems. Biochem Genet 2024; 62:18-39. [PMID: 37394575 DOI: 10.1007/s10528-023-10416-7] [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: 04/23/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023]
Abstract
A new era of medical technology in cancer treatment is a directly specific modification of gene expression in tumor cells by nucleic acid delivery. Currently, the main challenge to achieving this goal is to find a non-toxic, safe, and effective strategy for gene transfer to cancer cells. Synthetic composites based on cationic polymers have historically been favored in bioengineering due to their ability to mimic bimolecular structures. Among them, polyethylenimines (PEIs) with superior properties such as a wide range of molecular weight and a flexible structure may propel the development of functional combinations in the biomedical and biomaterial fields. Here, in this review, we will focus on the recent progressions in the formulation optimization of PEI-based polyplex in gene delivery to treat cancer. Also, the effect of PEI's intrinsic characteristics such as structure, molecular weight, and positive charges which influence the gene delivery efficiency will be discussed.
Collapse
Affiliation(s)
- Nejad Mohammadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunology Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nashmin Fayazi Hosseini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hossein Nemati
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Gholam Ali Kardar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Immunology Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Xiong Z, Tong T, Xie Z, Yu S, Zhuang R, Jia Q, Peng S, Li B, Xie J, Li K, Wu J, Huang H. Delivery of gefitinib loaded nanoparticles for effectively inhibiting prostate cancer progression. Biomater Sci 2024; 12:650-659. [PMID: 38168678 DOI: 10.1039/d3bm01735d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Androgen deprivation therapy is administered to suppress the growth of prostate cancer (PCa). However, some cells continue to proliferate independent of hormones, leading to the development of castration-resistant prostate cancer (CRPC). Overexpression of the epidermal growth factor receptor (EGFR) has been observed in CRPC and is associated with an unfavorable prognosis. Gefitinib (GEF) is an EGFR inhibitor used to treat patients with CRPC. Nevertheless, some clinical studies have reported that gefitinib does not result in prostate-specific antigen (PSA) or objectively measurable CRPC reactions. This lack of response may be attributed to the limited solubility in water, high side effects, low tumor aggregation, and insufficient tumor-specific reactions of GEF. In order to tackle these obstacles, we present a practical and efficient approach to administer GEF, encompassing the utilization of biocompatible nanostructures as a vehicle for drug delivery to augment its bioaccessibility and curative potency. Despite their small particle size, poly(D,L-lactide-co-glycolide) acid nanoparticles (PLGA NPs) exhibit a high drug-loading capacity, low toxicity, biocompatibility, biodegradability, and minimal immunogenicity. The drug delivery efficiency can be improved by employing GEF@PLGA NPs, which could also enhance drug cytotoxicity and impede the advancement of prostate cancer. Moreover, through experiments in vivo, it has been verified that GEF@PLGA NPs exhibit selective accumulation in the tumor and effectively restrain tumor growth. Therefore, the GEF@PLGA NPs hold great promise for the treatment of PCa.
Collapse
Affiliation(s)
- Zhi Xiong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Tong Tong
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518057, China
| | - Zhaoxiang Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Shunli Yu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Ruilin Zhuang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Qiang Jia
- Guangzhou City Polytechnic, Guangzhou, 510520, China
| | - Shirong Peng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Bingheng Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Junjia Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Kaiwen Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Jun Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| |
Collapse
|
3
|
Zhang Q, Liu N, Wang J, Liu Y, Wang K, Zhang J, Pan X. The Recent Advance of Cell-Penetrating and Tumor-Targeting Peptides as Drug Delivery Systems Based on Tumor Microenvironment. Mol Pharm 2023; 20:789-809. [PMID: 36598861 DOI: 10.1021/acs.molpharmaceut.2c00629] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cancer has become the primary reason for industrial countries death. Although first-line treatments have achieved remarkable results in inhibiting tumors, they could have serious side effects because of insufficient selectivity. Therefore, specific localization of tumor cells is currently the main desire for cancer treatment. In recent years, cell-penetrating peptides (CPPs), as a kind of promising delivery vehicle, have attracted much attention because they mediate the high-efficiency import of large quantities of cargos in vivo and vitro. Unfortunately, the poor targeting of CPPs is still a barrier to their clinical application. In order to solve this problem, researchers use the various characteristics of tumor microenvironment and multiple receptors to improve the specificity toward tumors. This review focuses on the characteristics of the tumor microenvironment, and introduces the development of strategies and peptides based on these characteristics as drug delivery system in the tumor-targeted therapy.
Collapse
Affiliation(s)
- Qingqing Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Nanxin Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuying Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Kai Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
4
|
Yang L, Fang H, Jiang J, Sha Y, Zhong Z, Meng F. EGFR-targeted pemetrexed therapy of malignant pleural mesothelioma. Drug Deliv Transl Res 2021; 12:2527-2536. [PMID: 34802094 DOI: 10.1007/s13346-021-01094-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2021] [Indexed: 12/17/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a rare malignancy with poor prognosis, for which chemotherapy with pemetrexed (PEM) is among the few clinical treatments. PEM suffers, however, fast clearance, moderate drug exposure, and dose-limiting toxicities. Here, we report on epidermal growth factor receptor (EGFR)-targeted disulfide-crosslinked biodegradable chimaeric polymersomes (EGFR-CPs) to firmly load PEM and boost chemotherapy of MPM. EGFR-CPs encapsulating 8.7-16.4 wt.% PEM (EGFR-CPs-PEM) showed diameters of 62-65 nm and reduction-responsive drug release property. EGFR-CPs-PEM was more efficiently taken up by EGFR-overexpressed MSTO-211H cells, inducing about 4.7-fold enhanced anticancer activity compared with non-targeted CPs-PEM control. Intriguingly, the in vivo experiments in MSTO-211H xenograft mouse model revealed that EGFR-CPs-PEM brought about superior tumor deposition and penetration to CPs-PEM, and significantly more potent tumor repression than CPs-PEM and free PEM. This polymersome-enabled EGFR-targeted delivery of PEM offers an appealing therapeutic strategy for MPM.
Collapse
Affiliation(s)
- Liang Yang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Hanghang Fang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Jingjing Jiang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Yongjie Sha
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China.
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China.
| |
Collapse
|
5
|
Gupta SS, Mishra V, Mukherjee MD, Saini P, Ranjan KR. Amino acid derived biopolymers: Recent advances and biomedical applications. Int J Biol Macromol 2021; 188:542-567. [PMID: 34384802 DOI: 10.1016/j.ijbiomac.2021.08.036] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 01/19/2023]
Abstract
Over the past few years, amino acids (AA) have emerged as promising biomaterials for the synthesis of functional polymers. Owing to the diversity of functional groups in amino acids, various polymerization methods may be used to make a wide range of well-defined functional amino-acid/peptide-based optically active polymers with varying polymer lengths, compositions, and designs. When incorporated with chirality and self-assembly, they offer a wide range of applications and are particularly appealing in the field of drug delivery, tissue engineering, and biosensing. There are several classes of these polymers that include polyamides (PA), polyesters (PE), poly(ester-amide)s (PEA)s, polyurethanes (PU)s, poly(depsipeptide)s (PDP)s, etc. They offer the ability to control functionality, conjugation, crosslinking, stimuli responsiveness, and tuneable mechanical/thermal properties. In this review, we present the recent advancements in the synthesis strategies for obtaining these amino acid-derived bio-macromolecules, their self-assembly properties, and the wealth of prevalent applications.
Collapse
Affiliation(s)
| | - Vivek Mishra
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, NOIDA, India.
| | | | | | - Kumar Rakesh Ranjan
- Amity Institute of Applied Sciences, Amity University Uttar Pradesh, NOIDA, India.
| |
Collapse
|
6
|
Liu X, Wu F, Cai K, Zhao Z, Zhang Z, Chen Y, Liu Y, Cheng J, Yin L. Cancer cell-targeted cisplatin prodrug delivery in vivo via metabolic labeling and bioorthogonal click reaction. Biomater Sci 2021; 9:1301-1312. [PMID: 33350407 DOI: 10.1039/d0bm01709d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The discrepancy of surface receptors on cancerous and non-cancerous cells has been regarded as the mainstay of cancer-targeted therapy. However, due to the heterogeneity of tumor cells and the insufficient levels of receptors on the tumor cell surface, the success of cancer cell-targeted therapies is largely limited. Histone deacetylase/cathepsin l-responsive acetylated azidomannose (DCL-AAM) was previously developed to effectively and selectively label cancer cell surfaces with reactive azido groups via sugar metabolism. Herein, the labeling kinetics and generality of DCL-AAM were systematically investigated in varieties of tumor cells in vitro and in SKOV3 xenograft tumors in vivo. Based on this, dibenzocyclooctyne-cisplatin (DBCO-Pt) prodrug was developed, and DCL-AAM-mediated metabolic labeling of SKOV3 cells enhanced the tumor accumulation of DBCO-Pt ∼2 fold via bioorthogonal click chemistry, potentiating the anti-tumor efficacy of cisplatin yet alleviating the systemic toxicity. This work, therefore, provides the experimental and theoretical support for the future design of sugar metabolism-based targeted delivery systems and may provide a promising candidate for the treatment of cancers lacking appropriate biomarkers.
Collapse
Affiliation(s)
- Xun Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Fan Wu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Kaimin Cai
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Ziyin Zhao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Zhimin Zhang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Yongbing Chen
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Yong Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| |
Collapse
|
7
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
8
|
In Situ Crosslinked Hydrogel Depot for Sustained Antibody Release Improves Immune Checkpoint Blockade Cancer Immunotherapy. NANOMATERIALS 2021; 11:nano11020471. [PMID: 33673289 PMCID: PMC7918828 DOI: 10.3390/nano11020471] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/06/2021] [Accepted: 02/09/2021] [Indexed: 01/22/2023]
Abstract
The therapeutic inhibition of immune checkpoints, including cytotoxic T lymphocyte-associated protein (CTLA)-4 and programmed cell death 1 (PD-1), through the use of function blocking antibodies can confer improved clinical outcomes by invigorating CD8+ T cell-mediated anticancer immunity. However, low rates of patient responses and the high rate of immune-related adverse events remain significant challenges to broadening the benefit of this therapeutic class, termed immune checkpoint blockade (ICB). To overcome these significant limitations, controlled delivery and release strategies offer unique advantages relevant to this therapeutic class, which is typically administered systemically (e.g., intravenously), but more recently, has been shown to be highly efficacious using locoregional routes of administration. As such, in this paper, we describe an in situ crosslinked hydrogel for the sustained release of antibodies blocking CTLA-4 and PD-1 signaling from a locoregional injection proximal to the tumor site. This formulation results in efficient and durable anticancer effects with a reduced systemic toxicity compared to the bolus delivery of free antibody using an equivalent injection route. This formulation and strategy thus represent an approach for achieving the efficient and safe delivery of antibodies for ICB cancer immunotherapy.
Collapse
|
9
|
Paiva I, Mattingly S, Wuest M, Leier S, Vakili MR, Weinfeld M, Lavasanifar A, Wuest F. Synthesis and Analysis of 64Cu-Labeled GE11-Modified Polymeric Micellar Nanoparticles for EGFR-Targeted Molecular Imaging in a Colorectal Cancer Model. Mol Pharm 2020; 17:1470-1481. [DOI: 10.1021/acs.molpharmaceut.9b01043] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Igor Paiva
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Stephanie Mattingly
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Melinda Wuest
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Canada
| | - Samantha Leier
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Mohammad Reza Vakili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Michael Weinfeld
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
| | - Frank Wuest
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Canada
| |
Collapse
|
10
|
Li C, Cai G, Song D, Gao R, Teng P, Zhou L, Ji Q, Sui H, Cai J, Li Q, Wang Y. Development of EGFR-targeted evodiamine nanoparticles for the treatment of colorectal cancer. Biomater Sci 2019; 7:3627-3639. [PMID: 31328737 DOI: 10.1039/c9bm00613c] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Invasion and metastasis of colorectal cancer (CRC) are leading causes of death of CRC patients. Previous findings demonstrate that evodiamine (Evo), an indolequinone alkaloid, is effective in combating CRC; however, its poor aqueous solubility and low oral bioavailability limit its application in the prevention of invasion and metastasis of CRC. It is known that selectively targeting cancer-specific receptors highly expressed on the surface of cancer cells by nanocarriers loaded with cytotoxic drugs is a viable strategy in nanobiotechnology to enhance cancer cell killing and minimize side effects. In this study, we report the development of a new class of nanotherapeutics: EGFR-targeting Evo-encapsulated poly(amino acid) nanoparticles (GE11-Evo-NPs). These nanoparticles exhibited good aqueous solubility, slow release, and active targeting capability. Their inhibitory effect on human colon cancer cells and therapeutic efficacy against invasion and metastasis of CRC in nude mice were systematically investigated. Mechanisms of the GE11-Evo-NPs against EGFR mediated invasion and metastasis of CRC were also explored. Compared with free Evo, the GE11-Evo-NPs showed significantly increased cytotoxicity to colon cancer cells and potently inhibited CRC LoVo cell adhesion, invasion, and migration. The expression of EGFR, VEGF, and MMP proteins was dramatically down-regulated, which may partially account for their inhibition of invasion and metastasis of CRC. Moreover, in vivo studies show that the GE11-Evo-NPs exhibited much greater potency than other control groups in inhibiting CRC invasion and metastasis, tumor volume, and growth in nude mice, leading to a significantly prolonged tumor-bearing survival duration (P < 0.01).
Collapse
Affiliation(s)
- Chunpu Li
- Department of Medical Oncology & Cancer institute of medicine, Shuguang Hospital, Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Killian T, Buntz A, Herlet T, Seul H, Mundigl O, Längst G, Brinkmann U. Antibody-targeted chromatin enables effective intracellular delivery and functionality of CRISPR/Cas9 expression plasmids. Nucleic Acids Res 2019; 47:e55. [PMID: 30809660 PMCID: PMC6547418 DOI: 10.1093/nar/gkz137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/21/2019] [Accepted: 02/20/2019] [Indexed: 01/01/2023] Open
Abstract
We report a novel system for efficient and specific targeted delivery of large nucleic acids to and into cells. Plasmid DNA and core histones were assembled to chromatin by salt gradient dialysis and subsequently connected to bispecific antibody derivatives (bsAbs) via a nucleic acid binding peptide bridge. The resulting reconstituted vehicles termed 'plasmid-chromatin' deliver packaged nucleic acids to and into cells expressing antigens that are recognized by the bsAb, enabling intracellular functionality without detectable cytotoxicity. High efficiency of intracellular nucleic acid delivery is revealed by intracellular expression of plasmid encoded genes in most (∼90%) target cells to which the vehicles were applied under normal growth/medium conditions in nanomolar concentrations. Specific targeting, uptake and transgene expression depends on antibody-mediated cell surface binding: plasmid chromatin of identical composition but with non-targeting bsAbs or without bsAbs is ineffective. Examples that demonstrate applicability, specificity and efficacy of antibody-targeted plasmid chromatin include reporter gene constructs as well as plasmids that enable CRISPR/Cas9 mediated genome editing of target cells.
Collapse
Affiliation(s)
- Tobias Killian
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Annette Buntz
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Teresa Herlet
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Heike Seul
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Olaf Mundigl
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Gernot Längst
- Biochemistry III; Biochemistry Centre Regensburg (BCR), University of Regensburg, Regensburg, Germany
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| |
Collapse
|
12
|
Jung S, Kim J, Pramanick S, Park H, Lee H, Lee J, Kim WJ. A Pt(iv)-mediated polymer architecture for facile and stimuli-responsive intracellular gene silencing with chemotherapy. Biomater Sci 2019; 6:3345-3355. [PMID: 30357140 DOI: 10.1039/c8bm01019f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Conventional chemotherapy has been impeded by the inherent characteristics of cancer including fast mutagenesis and drug resistance; thus a combination therapy consisting of multiple therapeutic strategies has attracted much attention. However, the loading processes of multiple therapeutic molecules affect each other; thus the development of a nanocarrier that enables independent loading of the cargo molecules has been demanded. Herein, we report an ingeniously designed Pt(iv)-mediated polymeric architecture (Pt-PA) for combinatorial gene and chemotherapy to address the issue, prepared by crosslinking a cationic polymer (polyethylenimine, PEI) with a Pt(iv) prodrug. Therapeutic siRNA (anti-BCL2) was simply loaded by electrostatic interaction to form a stable nanocomplex. In the cellular study, the simultaneous release of both the active Pt(ii) drug and siRNA was monitored under the intracellular reducing environment, driven by dissociation of the polymer architecture due to an inherent characteristic of the Pt(iv) crosslinker. Therefore, an enhanced gene silencing effect and an anticancer effect were observed. Furthermore, in the animal study, an improved therapeutic effect of the nanocomplex was observed, which can be explained by tumor targeting via the EPR effect, and enhanced drug and siRNA release at the intracellular environment simultaneously. Taken together, the overall results from in vitro and in vivo studies strongly suggest the therapeutic potential of our precisely designed Pt(iv)-mediated polymer architecture.
Collapse
Affiliation(s)
- Sungjin Jung
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.
| | | | | | | | | | | | | |
Collapse
|
13
|
Lieser RM, Chen W, Sullivan MO. Controlled Epidermal Growth Factor Receptor Ligand Display on Cancer Suicide Enzymes via Unnatural Amino Acid Engineering for Enhanced Intracellular Delivery in Breast Cancer Cells. Bioconjug Chem 2019; 30:432-442. [PMID: 30615416 DOI: 10.1021/acs.bioconjchem.8b00783] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Proteins are ideal candidates for disease treatment because of their high specificity and potency. Despite this potential, delivery of proteins remains a significant challenge due to the intrinsic size, charge, and stability of proteins. Attempts to overcome these challenges have most commonly relied on direct conjugation of polymers and peptides to proteins via reactive groups on naturally occurring residues. While such approaches have shown some success, they allow limited control of the spacing and number of moieties coupled to proteins, which can hinder bioactivity and delivery capabilities of the therapeutic. Here, we describe a strategy to site-specifically conjugate delivery moieties to therapeutic proteins through unnatural amino acid (UAA) incorporation, in order to explore the effect of epidermal growth factor receptor (EGFR)-targeted ligand valency and spacing on internalization of proteins in EGFR-overexpressing inflammatory breast cancer (IBC) cells. Our results demonstrate the ability to enhance targeted protein delivery by tuning a small number of EGFR ligands per protein and clustering these ligands to promote multivalent ligand-receptor interactions. Furthermore, the tailorability of this simple approach was demonstrated through IBC-targeted cell death via the delivery of yeast cytosine deaminase (yCD), a prodrug converting enzyme.
Collapse
Affiliation(s)
- Rachel M Lieser
- Department of Chemical and Biomolecular Engineering , University of Delaware , 150 Academy Street , Newark , Delaware 19716 , United States
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering , University of Delaware , 150 Academy Street , Newark , Delaware 19716 , United States
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering , University of Delaware , 150 Academy Street , Newark , Delaware 19716 , United States
| |
Collapse
|
14
|
Dai Y, Zhang X. MicroRNA Delivery with Bioreducible Polyethylenimine as a Non‐Viral Vector for Breast Cancer Gene Therapy. Macromol Biosci 2019; 19:e1800445. [DOI: 10.1002/mabi.201800445] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/13/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Yu Dai
- Engineering Research Center of Nano‐Geomaterials of Ministry of EducationFaculty of Materials Science and ChemistryChina University of Geosciences Wuhan 430074 China
| | - Xiaojin Zhang
- Engineering Research Center of Nano‐Geomaterials of Ministry of EducationFaculty of Materials Science and ChemistryChina University of Geosciences Wuhan 430074 China
| |
Collapse
|
15
|
Abstract
Gene therapy as a strategy for disease treatment requires safe and efficient gene delivery systems that encapsulate nucleic acids and deliver them to effective sites in the cell.
Collapse
Affiliation(s)
- Ziyao Kang
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| | - Qingbin Meng
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| | - Keliang Liu
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| |
Collapse
|
16
|
Sun H, Dong Y, Feijen J, Zhong Z. Peptide-decorated polymeric nanomedicines for precision cancer therapy. J Control Release 2018; 290:11-27. [DOI: 10.1016/j.jconrel.2018.09.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/27/2018] [Accepted: 09/30/2018] [Indexed: 01/12/2023]
|
17
|
Exploring pitfalls of 64Cu-labeled EGFR-targeting peptide GE11 as a potential PET tracer. Amino Acids 2018; 50:1415-1431. [PMID: 30039310 DOI: 10.1007/s00726-018-2616-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/09/2018] [Indexed: 12/29/2022]
Abstract
The epidermal growth factor receptor (EGFR) represents an important molecular target for both radiotracer-based diagnostic imaging and radionuclide therapy of various cancer entities. For the delivery of radionuclides to the tumor, peptides hold great potential as a transport vehicle. With respect to EGFR, the peptide YHWYGYTPQNVI (GE11) has been reported to bind the receptor with high specificity and affinity. In the present study, GE11 with β-alanine (β-Ala-GE11) was conjugated to the chelating agent p-SCN-Bn-NOTA and radiolabeled with 64Cu for the first radio pharmacological evaluation as a potential probe for positron emission tomography (PET)-based cancer imaging. For better water solubility, an ethylene glycol-based linker was introduced between the peptide's N terminus and the radionuclide chelator. The stability of the 64Cu-labeled peptide conjugate and its binding to EGFR-expressing tumor cells was investigated in vitro and in vivo, and then compared with the 64Cu-labeled EGFR-targeting antibody conjugate NOTA-cetuximab. The GE11 peptide conjugate [64Cu]Cu-NOTA-linker-β-Ala-GE11 ([64Cu]Cu-1) was stable in a buffer solution for at least 24 h but only 50% of the original compound was detected after 24 h of incubation in human serum. Stability could be improved by amidation of the peptide's C terminus (β-Ala-GE11-NH2 (2)). Binding assays with both conjugates, [64Cu]Cu-1 and [64Cu]Cu-2, using the EGFR-expressing tumor cell lines A431 and FaDu showed no specific binding. A pilot small animal PET investigation in FaDu tumor-bearing mice revealed only low tumor uptake (standard uptake value (SUV) < 0.2) for both conjugates. The best tumor-to-muscle ratio determined was 3.75 for [64Cu]Cu-1, at 1 h post injection. In conclusion, the GE11 conjugates in its present form are not suitable for further biological investigations, since they presumably form aggregates.
Collapse
|
18
|
Wang C, Liu L, Cao H, Zhang W. Intracellular GSH-activated galactoside photosensitizers for targeted photodynamic therapy and chemotherapy. Biomater Sci 2018; 5:274-284. [PMID: 27942618 DOI: 10.1039/c6bm00482b] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ligand-targeted cancer therapeutics has been developed to minimize non-specific cytotoxicity via ligand-drug conjugates during the past few decades. We present here the design and synthesis of a GSH-activated amphiphilic photosensitizer conjugated with galactose (TPP-S-S-Gal) for targeted photodynamic therapy. Furthermore, the galactoside photosensitizer as supramolecular amphiphiles can self-assemble into micelles, which can be applied in integrative cancer treatment with chemotherapy drugs such as camptothecin (CPT) encapsulated in the hydrophobic core of micelles. Upon reaction with free thiol GSH that is relatively abundant in tumor cells, disulfide bond cleavage occurs as well as the active photosensitizer TPP and chemotherapy drug CPT release, which can cause cell apoptosis. The in vitro biological assessment of TPP-S-S-Gal micelles against the A549 cell line was evaluated by MTT assay, flow cytometry and confocal scanning laser microscopy, respectively. According to the MTT assay, TPP-S-S-Gal micelles exhibited low dark toxicity and efficient integrative efficacy of PDT and chemotherapy towards A549 cells after light irradiation.
Collapse
Affiliation(s)
- Chaochao Wang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Lichao Liu
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Hongliang Cao
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Weian Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| |
Collapse
|
19
|
Alazzo A, Lovato T, Collins H, Taresco V, Stolnik S, Soliman M, Spriggs K, Alexander C. Structural variations in hyperbranched polymers prepared via thermal polycondensation of lysine and histidine and their effects on DNA delivery. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/jin2.36] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Ali Alazzo
- School of Pharmacy; University of Nottingham; Nottingham NG7 2RD UK
- Department of Pharmaceutics; University of Mosul; Mosul Iraq
| | - Tatiana Lovato
- School of Pharmacy; University of Nottingham; Nottingham NG7 2RD UK
| | - Hilary Collins
- School of Pharmacy; University of Nottingham; Nottingham NG7 2RD UK
| | - Vincenzo Taresco
- School of Pharmacy; University of Nottingham; Nottingham NG7 2RD UK
| | - Snjezana Stolnik
- School of Pharmacy; University of Nottingham; Nottingham NG7 2RD UK
| | - Mahmoud Soliman
- Department of Pharmaceutics; Ain Shams University; Cairo Egypt
| | - Keith Spriggs
- School of Pharmacy; University of Nottingham; Nottingham NG7 2RD UK
| | | |
Collapse
|
20
|
Sun Y, Liu H, Cheng L, Zhu S, Cai C, Yang T, Yang L, Ding P. Thiol Michael addition reaction: a facile tool for introducing peptides into polymer-based gene delivery systems. POLYM INT 2017. [DOI: 10.1002/pi.5490] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Yanping Sun
- School of Pharmacy; Shenyang Pharmaceutical University; Shenyang China
| | - Hui Liu
- School of Pharmacy; Shenyang Pharmaceutical University; Shenyang China
| | - Lin Cheng
- School of Pharmacy; Shenyang Pharmaceutical University; Shenyang China
| | - Shimeng Zhu
- School of Pharmacy; Shenyang Pharmaceutical University; Shenyang China
| | - Cuifang Cai
- School of Pharmacy; Shenyang Pharmaceutical University; Shenyang China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences; Husson University; Bangor ME USA
| | - Li Yang
- School of Pharmacy; Shenyang Pharmaceutical University; Shenyang China
| | - Pingtian Ding
- School of Pharmacy; Shenyang Pharmaceutical University; Shenyang China
| |
Collapse
|
21
|
Oroojalian F, Rezayan AH, Mehrnejad F, Nia AH, Shier WT, Abnous K, Ramezani M. Efficient megalin targeted delivery to renal proximal tubular cells mediated by modified-polymyxin B-polyethylenimine based nano-gene-carriers. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017. [DOI: 10.1016/j.msec.2017.05.068] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
22
|
Sun Y, Yang Z, Wang C, Yang T, Cai C, Zhao X, Yang L, Ding P. Exploring the role of peptides in polymer-based gene delivery. Acta Biomater 2017; 60:23-37. [PMID: 28778533 DOI: 10.1016/j.actbio.2017.07.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/14/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022]
Abstract
Polymers are widely studied as non-viral gene vectors because of their strong DNA binding ability, capacity to carry large payload, flexibility of chemical modifications, low immunogenicity, and facile processes for manufacturing. However, high cytotoxicity and low transfection efficiency substantially restrict their application in clinical trials. Incorporating functional peptides is a promising approach to address these issues. Peptides demonstrate various functions in polymer-based gene delivery systems, such as targeting to specific cells, breaching membrane barriers, facilitating DNA condensation and release, and lowering cytotoxicity. In this review, we systematically summarize the role of peptides in polymer-based gene delivery, and elaborate how to rationally design polymer-peptide based gene delivery vectors. STATEMENT OF SIGNIFICANCE Polymers are widely studied as non-viral gene vectors, but suffer from high cytotoxicity and low transfection efficiency. Incorporating short, bioactive peptides into polymer-based gene delivery systems can address this issue. Peptides demonstrate various functions in polymer-based gene delivery systems, such as targeting to specific cells, breaching membrane barriers, facilitating DNA condensation and release, and lowering cytotoxicity. In this review, we highlight the peptides' roles in polymer-based gene delivery, and elaborate how to utilize various functional peptides to enhance the transfection efficiency of polymers. The optimized peptide-polymer vectors should be able to alter their structures and functions according to biological microenvironments and utilize inherent intracellular pathways of cells, and consequently overcome the barriers during gene delivery to enhance transfection efficiency.
Collapse
Affiliation(s)
- Yanping Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhen Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chunxi Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME, USA
| | - Cuifang Cai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaoyun Zhao
- Department of Microbiology and Cell Biology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Li Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
23
|
Chen J, Guan X, Hu Y, Tian H, Chen X. Peptide-Based and Polypeptide-Based Gene Delivery Systems. Top Curr Chem (Cham) 2017; 375:32. [DOI: 10.1007/s41061-017-0115-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/28/2017] [Indexed: 12/15/2022]
|
24
|
Jang D, Lee YM, Lee J, Doh J, Kim WJ. Remission of lymphoblastic leukaemia in an intravascular fluidic environment by pliable drug carrier with a sliding target ligand. Sci Rep 2017; 7:40739. [PMID: 28094326 PMCID: PMC5240144 DOI: 10.1038/srep40739] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/09/2016] [Indexed: 12/14/2022] Open
Abstract
A polyrotaxane-based nanoconstruct with pliable structure carrying a chemotherapeutic drug was developed for targeting circulating lymphoblastic leukaemia cells in a fluidic environment of blood vessels in vivo. By introducing lymphoblast targeting aptamer DNA through cyclodextrin, threaded in poly(ethylene glycol) as polyrotaxane, target aptamer slides along the long polymeric chain and actively search for target ligand, leading to active targeting in dynamic fluidic system which is enhanced by up to 6–fold compared with that of control carriers with non–sliding targeting ligands. Moreover, the drug carrier was made stimuli-responsive by employing i-motif DNA to selective releases of its payload at intracellular acidic condition. These combined features resulted in the effective remission of lymphoblastic leukaemia both in vitro and in dynamic blood vessels in vivo.
Collapse
Affiliation(s)
- Donghyun Jang
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS), Pohang 37673, Republic of Korea.,Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Yeong Mi Lee
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS), Pohang 37673, Republic of Korea.,Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Jaehyun Lee
- School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Junsang Doh
- School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea.,Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Won Jong Kim
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS), Pohang 37673, Republic of Korea.,Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| |
Collapse
|
25
|
Li J, Mao H, Kawazoe N, Chen G. Insight into the interactions between nanoparticles and cells. Biomater Sci 2017; 5:173-189. [DOI: 10.1039/c6bm00714g] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review summarizes the latest advances in nanoparticle (NP)–cell interactions. The influence of NP size, shape, shell structure, surface chemistry and protein corona formation on cellular uptake and cytotoxicity is highlighted in detail. Their impact on other cellular responses such as cell proliferation, differentiation and cellular mechanics is also discussed.
Collapse
Affiliation(s)
- Jingchao Li
- International Center for Materials Nanoarchitectonics
- National Institute for Materials Science
- Tsukuba
- Japan
- Department of Materials Science and Engineering
| | - Hongli Mao
- International Center for Materials Nanoarchitectonics
- National Institute for Materials Science
- Tsukuba
- Japan
- Department of Materials Science and Engineering
| | - Naoki Kawazoe
- International Center for Materials Nanoarchitectonics
- National Institute for Materials Science
- Tsukuba
- Japan
| | - Guoping Chen
- International Center for Materials Nanoarchitectonics
- National Institute for Materials Science
- Tsukuba
- Japan
- Department of Materials Science and Engineering
| |
Collapse
|
26
|
Deng Q, Li X, Zhu L, He H, Chen D, Chen Y, Yin L. Serum-resistant, reactive oxygen species (ROS)-potentiated gene delivery in cancer cells mediated by fluorinated, diselenide-crosslinked polyplexes. Biomater Sci 2017; 5:1174-1182. [DOI: 10.1039/c7bm00334j] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Fluorinated, diselenide-crosslinked polyplexes were developed to enable ROS-responsive and serum-resistant gene delivery in cancer cells.
Collapse
Affiliation(s)
- Qiurong Deng
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- P.R. China
| | - Xudong Li
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- P.R. China
| | - Lipeng Zhu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- P.R. China
| | - Hua He
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- P.R. China
| | - Donglai Chen
- Department of Thoracic Surgery
- Shanghai Pulmonary Hospital
- Tongji University School of Medicine
- Shanghai
- P.R. China
| | - Yongbing Chen
- Department of Cardiothoracic Surgery
- The Second Affiliated Hospital of Soochow University
- Suzhou 215004
- P.R. China
| | - Lichen Yin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- P.R. China
| |
Collapse
|
27
|
Liu J, Xu L, Jin Y, Qi C, Li Q, Zhang Y, Jiang X, Wang G, Wang Z, Wang L. Cell-Targeting Cationic Gene Delivery System Based on a Modular Design Rationale. ACS APPLIED MATERIALS & INTERFACES 2016; 8:14200-14210. [PMID: 27191222 DOI: 10.1021/acsami.6b04462] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
En route to target cells, a gene carrier faces multiple extra- and intracellular hurdles that would affect delivery efficacy. Although diverse strategies have been proposed to functionalize gene carriers for individually overcoming these barriers, it is challenging to generate a single multifunctional gene carrier capable of surmounting all these barriers. Aiming at this challenge, we have developed a supramolecular modular approach to fabricate a multifunctional cationic gene delivery system. It consists of two prefunctionalized modules: (1) a host module: a polymer (PCD-SS-PDMAEMA) composed of poly(β-cyclodextrin) backbone and disulfide-linked PDMAEMA arms, expectedly acting to compact DNA and release DNA upon cleavage of disulfide linkers in reductive microenvironment; and (2) a guest module: adamantyl and folate terminated PEG (Ad-PEG-FA), expectedly functioning to reduce nonspecific interactions, improve biocompatibility, and provide folate-mediated cellular targeting specificity. Through the host-guest interaction between β-cyclodextrin units of the "host" module and adamantyl groups of the "guest" module, the PCD-SS-PDMAEMA-1 (host) and Ad-PEG-FA (guest) self-assemble forming a supramolecular pseudocopolymer (PCD-SS-PDMAEMA-1/PEG-FA). Our comprehensive analyses demonstrate that the functions preassigned to the two building modules are well realized. The gene carrier effectively compacts DNA into stable nanosized polyplexes resistant to enzymatic digestion, triggers DNA release in reducing environment, possesses significantly improved hemocompatibility, and specifically targets folate-receptor positive cells. Most importantly, endowed with these predesigned functions, the PCD-SS-PDMAEMA-1/PEG-FA supramolecular gene carrier exhibits excellent transfection efficacy for both pDNA and siRNA. Thus, this work represents a proof-of-concept example showing the efficiency and convenience of an adaptable, modular approach for conferring multiple functions to a single supramolecular gene carrier toward effective in vivo delivery of therapeutic nucleic acids.
Collapse
Affiliation(s)
- Jia Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Luming Xu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yang Jin
- Department of Respiration, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei 430022, China
| | - Chao Qi
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Qilin Li
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yunti Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University , Wuhan 430072, China
| | - Xulin Jiang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University , Wuhan 430072, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei 430022, China
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei 430022, China
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei 430022, China
| |
Collapse
|