1
|
Yao Z, Liu T, Wang J, Fu Y, Zhao J, Wang X, Li Y, Yang X, He Z. Targeted delivery systems of siRNA based on ionizable lipid nanoparticles and cationic polymer vectors. Biotechnol Adv 2025; 81:108546. [PMID: 40015385 DOI: 10.1016/j.biotechadv.2025.108546] [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: 09/23/2024] [Revised: 02/04/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
As an emerging therapeutic tool, small interfering RNA (siRNA) had the capability to down-regulate nearly all human mRNAs via sequence-specific gene silencing. Numerous studies have demonstrated the substantial potential of siRNA in the treatment of broad classes of diseases. With the discovery and development of various delivery systems and chemical modifications, six siRNA-based drugs have been approved by 2024. The utilization of siRNA-based therapeutics has significantly propelled efforts to combat a wide array of previously incurable diseases and advanced at a rapid pace, particularly with the help of potent targeted delivery systems. Despite encountering several extracellular and intracellular challenges, the efficiency of siRNA delivery has been gradually enhanced. Currently, targeted strategies aimed at improving potency and reducing toxicity played a crucial role in the druggability of siRNA. This review focused on recent advancements on ionizable lipid nanoparticles (LNPs) and cationic polymer (CP) vectors applied for targeted siRNA delivery. Based on various types of targeted modifications, we primarily described delivery systems modified with receptor ligands, peptides, antibodies, aptamers and amino acids. Finally, we discussed the challenges and opportunities associated with siRNA delivery systems based on ionizable LNPs and CPs vectors.
Collapse
Affiliation(s)
- Ziying Yao
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Taiqing Liu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingwen Wang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunhai Fu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinhua Zhao
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoyu Wang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yinqi Li
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaodong Yang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyao He
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Xu L, Li S, Wan S, Liu Z, Zhong Y, Qian X, Qin J, Cai L, Huang H. Poly-lysine-modified recombinant protein nanocages for effective delivery of small activating RNA. J Control Release 2025; 382:113638. [PMID: 40139394 DOI: 10.1016/j.jconrel.2025.113638] [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: 11/10/2024] [Revised: 01/26/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
Small activating RNA (saRNA) holds significant promise as a therapeutic platform for various diseases. However, the development of efficient nanocarriers that can overcome existing delivery challenges and ensure effective cellular uptake remains a critical hurdle. In this study, we aimed to address this issue by genetically modifying four lysine residues at the N-terminus of the FTH gene through gene recombinant technology, resulting in the creation of poly-lysine-H-apoferrin (4LF) vectors. These vectors were designed to efficiently deliver saRNA encoding the Sirtuin1 (Sirt1) protein to chondrocytes, thereby mitigating cartilage damage. The poly-lysine modification conferred the ability of 4LF@saRNA nanoparticles (NPs) to escape from lysosomes via proton sponge effects and to release saRNA into the cytoplasm through the pH-induced degradation of the 4LF vector, ultimately activating the target gene. To enhance the retention of NPs within the joint cavity and facilitate intra-articular delivery, we incorporated the 4LF@saRNA NPs into a thermosensitive self-healing hydrogel composed of chitosan, oxidized chondroitin sulfate (OCS), and sodium β-glycerophosphate (β-GP). Experimental results demonstrated that the chitosan/OCS/β-GP-4LF@saRNA (OCCG-4LF@saRNA) delivery system effectively delivered the 4LF@saRNA NPs to chondrocytes both in vitro and in vivo, resulting in a significant increase in Sirt1 protein expression. This upregulation led to a reduction in chondrocyte apoptosis, enhanced cell migration, and improved cartilage protection, effectively alleviating symptoms of osteoarthritis. In conclusion, our findings suggested that the 4LF-based delivery system hold considerable potential for effective intracellular saRNA delivery, demonstrating promising biocompatibility, stability, and delivery efficiency, with significant therapeutic implications for cartilage repair.
Collapse
Affiliation(s)
- Lixing Xu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Shanshan Li
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Shen Wan
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Zhikuan Liu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yuxin Zhong
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Xiaoyang Qian
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Jiayi Qin
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Liangliang Cai
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China.
| | - Haiqin Huang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China.
| |
Collapse
|
3
|
Liu Y, Ma H, Zhang Q, Zhu Y, Chen L, Wang B, Cheng C, Lu F, Jiang T. Caffeic acid borate functionalized linear polyglycidol for targeted siRNA delivery in the treatment of primary biliary cholangitis. Int J Biol Macromol 2025; 311:143673. [PMID: 40334881 DOI: 10.1016/j.ijbiomac.2025.143673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025]
Abstract
Interfering with protein arginine methyltransferase 5 (PRMT5) gene expression with siRNA targeting is a promising strategy for precision primary biliary cholangitis (PBC) therapy, yet achieving effective and specific delivery of therapeutic agents to the liver remains challenging. In this study, we develop a caffeic acid borate derived linear polyglycidol polymer micelle CAPG, which can efficiently deliver siRNA to liver. The degradation of borate and hydrazone bonds in the high-ROS and acidic environment of inflammatory cells triggers micelle disintegration, facilitating siRNA release. Comprehensive in vivo experiments demonstrate that siRNA1@CAPG can significantly reduce the concentration of liver inflammation-related biomarkers and pro-inflammatory factors and notably increase superoxide dismutase (SOD) levels in mice, outperforming free siRNA1 in suppressing PRMT5 expression. Therefore, our study offers valuable reference for the treatment of primary biliary cholangitis.
Collapse
Affiliation(s)
- Ying Liu
- Affiliated Changshu Hospital of Nantong University, No. 18 Taishan Road, Changshu City, Jiangsu Province, China
| | - Haixia Ma
- School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Qiange Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuehong Zhu
- Affiliated Changshu Hospital of Nantong University, No. 18 Taishan Road, Changshu City, Jiangsu Province, China
| | - Liangyun Chen
- Affiliated Changshu Hospital of Nantong University, No. 18 Taishan Road, Changshu City, Jiangsu Province, China
| | - Bing Wang
- Affiliated Changshu Hospital of Nantong University, No. 18 Taishan Road, Changshu City, Jiangsu Province, China
| | - Cuie Cheng
- Affiliated Changshu Hospital of Nantong University, No. 18 Taishan Road, Changshu City, Jiangsu Province, China
| | - Fenying Lu
- Affiliated Changshu Hospital of Nantong University, No. 18 Taishan Road, Changshu City, Jiangsu Province, China
| | - Tingwang Jiang
- Affiliated Changshu Hospital of Nantong University, No. 18 Taishan Road, Changshu City, Jiangsu Province, China.
| |
Collapse
|
4
|
Ma Y, Fung V, VanKeulen-Miller R, Tiwade PB, Narasipura EA, Gill NA, Fenton OS. A Metabolite Co-Delivery Strategy to Improve mRNA Lipid Nanoparticle Delivery. ACS APPLIED MATERIALS & INTERFACES 2025; 17:26202-26215. [PMID: 40274610 DOI: 10.1021/acsami.4c22969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Lipid nanoparticles (LNPs) effectively protect mRNA and facilitate its entry into target cells for protein synthesis. Despite these successes, cellular entry alone may not be enough for optimal protein expression, as mRNA translation also depends on the availability of essential metabolites, including metabolic energy sources, coenzymes, and amino acids. Without adequate metabolites, mRNA translation may be less efficient, potentially leading to higher dosing requirements or poorer therapeutic outcomes for LNP therapies. To address this, we develop a metabolite co-delivery strategy by encapsulating essential metabolites within mRNA LNPs, hypothesizing that our approach can uniformly improve mRNA delivery. Instead of adding a fifth component to the organic phase, our strategy involves mixing the metabolite with the mRNA payload in the aqueous phase, while maintaining the molar ratio of the components in the organic phase during LNP formulation. We verify our approach in vitro and in vivo, highlighting the broad applicability of our strategy through mechanism and efficacy studies across multiple cell lines, and physiological conditions, such as normoxia (i.e., 21% oxygen), hypoxia (i.e., 1% oxygen), and in mice. Taken collectively, we anticipate that our metabolite co-delivery strategy may serve as a generalizable strategy to enhance in vitro and in vivo protein expression using mRNA LNPs, potentially offering broad applicability for the study and treatment of disease.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Vincent Fung
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Palas B Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Eshan A Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Nicole A Gill
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
5
|
Liu S, Wan X, Gou Y, Yang W, Xu W, Du Y, Peng X, Wang X, Zhang X. The emerging functions and clinical implications of circRNAs in acute myeloid leukaemia. Cancer Cell Int 2025; 25:167. [PMID: 40296024 PMCID: PMC12038945 DOI: 10.1186/s12935-025-03772-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Acute myeloid leukaemia (AML) is a prevalent haematologic malignancy characterized by significant heterogeneity. Despite the application of aggressive therapeutic approaches, AML remains associated with poor prognosis. Circular RNAs (circRNAs) constitute a unique class of single-stranded RNAs featuring covalently closed loop structures that are ubiquitous across species. These molecules perform crucial regulatory functions in the pathogenesis of various diseases through diverse mechanisms, including acting as miRNA sponges, interacting with DNA or proteins, and encoding functional proteins/polypeptides. Recently, numerous circRNAs have been confirmed to have aberrant expression patterns in AML patients. In particular, certain circRNAs are closely associated with specific clinicopathological characteristics and thus have great potential as diagnostic/prognostic biomarkers and therapeutic targets in AML. Herein, we systematically summarize the biogenesis, degradation, and functional mechanisms of circRNAs while highlighting their clinical relevance. We also outline a series of online databases and analytical tools available to facilitate circRNA research. Finally, we discuss the current challenges and future research priorities in this evolving field.
Collapse
Affiliation(s)
- Shuiqing Liu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Xingyu Wan
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Yang Gou
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Wuchen Yang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Wei Xu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Yuxuan Du
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Xiangui Peng
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400037, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
6
|
Cui C, Tang J, Chen J, Zhang B, Li R, Zhang Q, Qiu C, Chen R, Min G, Sun Z, Weng H. Lactobacillus acidophilus extracellular vesicles-coated UiO-66-NH 2@siRNA nanoparticles for ulcerative colitis targeted gene therapy and gut microbiota modulation. J Nanobiotechnology 2025; 23:301. [PMID: 40247297 PMCID: PMC12007195 DOI: 10.1186/s12951-025-03376-0] [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: 12/26/2024] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
Ulcerative colitis (UC) is a complex and chronic inflammatory bowel disease whose pathogenesis involves genetic and environmental factors, which poses a challenge for treatment. Here, we have designed an innovative integrated therapeutic strategy using Lactobacillus acidophilus extracellular vesicles (EVs) to encapsulate UiO-66-NH2 nanoparticles bounded with TNF-α siRNA (EVs@UiO-66-NH2@siRNA) for UC treatment. This system shows superior affinity to inflammation-related cells due to the Lactobacillus acidophilus EVs can maintain immune homeostasis by regulating the secretion of cytokines in vitro. siRNA can specifically target the key inflammatory TNF-α in UC and silence its gene expression, thereby regulating the process of inflammatory response. After oral administration, EVs@UiO-66-NH2@siRNA demonstrates an accurate delivery of TNF-α siRNA to colonize the colon site and exerts a siRNA therapeutic effect by inhibiting the expression of TNF-α, which alleviates the intestinal inflammation in DSS-induced UC model. Moreover, this system can modulate the types and compositional structures of gut microbiota and metabolites to achieve an anti-inflammatory phenotype, which is helpful for the repair of intestinal homeostasis. We also have proved that UiO-66-NH2 nanoparticles exhibit a high loading capacity for TNF-α siRNA and good pH responsiveness, improving the potent release of siRNA in colon tissue. Collectively, the EVs@UiO-66-NH2@siRNA nano-delivery system demonstrate a feasible combination therapeutic strategy for UC through gut microecology modulation, immune regulation and TNF-α siRNA silence, which may provide a potential targeted treatment approach for inflammatory bowel disease.
Collapse
Affiliation(s)
- Chenyang Cui
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Jiaze Tang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Jie Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Beining Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Ruonan Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Qiang Zhang
- Department of Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Chunjing Qiu
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Rongchen Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Geng Min
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zhaowei Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Haibo Weng
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
7
|
Yazdi M, Burghardt T, Seidl J, Lächelt U, Wagner E. Evolution of Lipo-Xenopeptide Carriers for siRNA Delivery: Interplay of Stabilizing Subunits. Bioconjug Chem 2025; 36:846-858. [PMID: 40134240 DOI: 10.1021/acs.bioconjchem.5c00096] [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: 03/27/2025]
Abstract
Although small interfering RNA (siRNA) holds immense promise for treating genetic diseases and cancers, its clinical application is constrained by instability, cellular uptake barriers, and inefficient cytosolic delivery, underscoring the need for effective delivery systems. Therefore, this study focuses on screening novel T-shaped lipo-xenopeptide (XP) nanocarriers for siRNA polyplex formulation, integrating two single succinoyl-tetraethylene pentamine (Stp) units for electrostatic interaction and tyrosine tripeptides (Y3) for aromatic stabilization, along with structural modifications such as the addition of histidine (H) with or without terminal cysteines (C), and the incorporation of various fatty acids (FAs). A systematic evaluation of siRNA binding, nanoparticle stability, and gene silencing efficiency in multiple cell lines illustrated that the novel Stp1-HC lipo-XPs carriers outperform their Stp2-HC analogs, despite having fewer cationizable Stp units. This advantage stems from increased fatty acid, Y3, and C density, which compensates for reduced electrostatic interactions. The presence of H in combination with unsaturated FAs significantly improved the functional siRNA delivery. Our findings highlight the complex interplay of electrostatic, hydrophobic, covalent, hydrogen-bonded, and aromatic interactions to achieve efficient siRNA delivery, which is best-balanced in the oleic acid-containing Stp1-HC/OleA lipo-XP, exceeding the previously best standard carrier Stp2-HC/OleA in efficiency.
Collapse
Affiliation(s)
- Mina Yazdi
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU), 81377 Munich, Germany
- CNATM - Cluster for Nucleic Acid Therapeutics, 81377 Munich, Germany
| | - Tobias Burghardt
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU), 81377 Munich, Germany
| | - Johanna Seidl
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU), 81377 Munich, Germany
| | - Ulrich Lächelt
- Center for Nanoscience (CeNS), Ludwig-Maximilians-Universität (LMU), 80539 Munich, Germany
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, Vienna 1090, Austria
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU), 81377 Munich, Germany
- CNATM - Cluster for Nucleic Acid Therapeutics, 81377 Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-Universität (LMU), 80539 Munich, Germany
| |
Collapse
|
8
|
Ebenezer O, Oyebamiji AK, Olanlokun JO, Tuszynski JA, Wong GKS. Recent Update on siRNA Therapeutics. Int J Mol Sci 2025; 26:3456. [PMID: 40331977 PMCID: PMC12026779 DOI: 10.3390/ijms26083456] [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: 02/01/2025] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
Small interfering RNA (siRNA) has been deemed a promising therapeutic method for treating diverse diseases. siRNA-based therapeutics provide a distinct mechanism of action by selectively targeting and silencing disease-causing genes at the post-transcriptional level. This paper provides an overview of the present state of siRNA-based therapeutics, highlighting their potential in different therapeutic areas. The first section of this review introduces the basic principles of siRNA technology, including its mechanism of action and delivery methods. Subsequently, we discuss the impediments associated with siRNA delivery and manufacturing development and the strategies for overcoming these obstacles. The clinical advancement of siRNA therapeutics in various disease areas, including cancer, genetic disorders, viral infections, and inflammatory diseases, is summarized. Lastly, we summarize the successes, failures, and lessons learned from the development of siRNAs. With advancements in delivery systems and improvements in target selection, the field of medicine can be revolutionized, and siRNA therapeutics can offer new treatment options for patients.
Collapse
Affiliation(s)
- Oluwakemi Ebenezer
- Department of Physics, University of Alberta, Edmonton, AB T6G 2E1, Canada;
| | | | - John Oludele Olanlokun
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan 200005, Nigeria;
| | - Jack A. Tuszynski
- Department of Physics, University of Alberta, Edmonton, AB T6G 2E1, Canada;
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Department of Mechanical and Aerospace Engineering (DIMEAS), Politecnico di Torino, 10129 Turin, Italy
- Department of Data Science and Engineering, The Silesian University of Technology, 44-100 Gliwice, Poland
| | - Gane Ka-Shu Wong
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada;
| |
Collapse
|
9
|
Kundapura S, Craske D, Hickman G, Braim S. Enhanced siRNA delivery with novel smart chitosan-based formulations. J Pharm Sci 2025; 114:103670. [PMID: 39914729 DOI: 10.1016/j.xphs.2025.01.006] [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: 08/25/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 02/17/2025]
Abstract
This study aims to develop an innovative multifunctional and dual responsive drug formulation for precise siRNA delivery to breast cancer sites, addressing the challenges posed by conventional cancer treatments which often result in adverse side effects due to their non-specific nature. The formulation made by incorporating gold coated superparamagnetic iron oxide nanoparticles (Au-SPIONs) into chitosan microspheres, which were subsequently loaded with siRNA. Comprehensive characterization, including scanning electron microscopy (SEM), transmission electron microscopy (TEM), dynamic light scattering (DLS), Fourier-transform infrared spectroscopy (FTIR), and energy-dispersive X-ray spectroscopy (EDS) confirmed the formulation's favourable morphology, particle size distribution, chemical composition, and stability, indicating its strong potential for effective siRNA drug delivery applications. The developed formulation demonstrated siRNA encapsulation efficiencies ranging from 27.4 % to 88.6 % and loading capacity from 0.291 % to 1.59 %, these values particularly higher for medium molecular weight chitosan. These results were compared across different formulations, showing that variations in chitosan type and crosslinker concentration significantly influenced encapsulation efficiency and drug release profiles. Additionally, our results were compared to previous studies on chitosan microspheres encapsulating organic drugs and siRNA, where the developed system demonstrated similar encapsulation and release properties.. The type of chitosan and the choice of crosslinker significantly influenced the drug release patterns. Diverse release profiles across batches highlighted the necessity for precise formulation control. Incorporating SPIONs into chitosan microspheres presents a promising strategy for magnetically driven, site-specific drug delivery. The dual pH-responsive and magnetic properties enable rapid and targeted siRNA release, leveraging the acidic tumor microenvironment as an internal stimulus in addition to external magnetic stimuli. This novel combination of SPIONs, chitosan microspheres, and siRNA encapsulation represents a new approach for targeted drug delivery. While further research is needed to refine and optimize this approach, our study provides a proof of concept for advancing targeted cancer therapies.
Collapse
Affiliation(s)
- Srujan Kundapura
- School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Dominic Craske
- School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Graham Hickman
- School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Shwana Braim
- School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom.
| |
Collapse
|
10
|
Su B, Chen Q, Li X, Fang M, Wang Y, Song H, You H, Zhou Z, Wu Y, Zhao Z, Chen Y, Fan H, Li C, Jiang C, Sun T. A Methionine Allocation Nanoregulator for the Suppression of Cancer Stem Cells and Support to the Immune Cells by Epigenetic Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415207. [PMID: 39985256 PMCID: PMC12005795 DOI: 10.1002/advs.202415207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/13/2025] [Indexed: 02/24/2025]
Abstract
Epigenetic dysregulation is prevalent in human cancers, affecting gene expression and metabolic patterns to meet the demands of malignant evolution and abnormal epigenetic processes, and resulting in a protumor immune microenvironment. Tumors require a steady supply of methionine for maintaining epigenetic flexibility, which is the only exogenous precursor of methyl donor S-adenosylmethionine for methylation, crucial for their resistance to therapies and survival in a nutrient-deficient microenvironment. Thus, tumor cells upregulate the Lat4 transporter to compete and deprive methionine in the microenvironment, sustaining their malignant phenotypes and also impairing immune cell functions. Addressing this methionine addiction is the key to overcoming drug resistance and improving immune response. Despite the challenge of lacking specific Lat4 inhibitors, an oxaliplatin prodrug crosslinked fluorinated polycation/anti-Lat4 small interfering RNA complex nanoregulator (AS-F-NP) has been designed and developed here. This nanoregulator restricted the greedy methionine uptake of tumor cells by knocking down Lat4, which in turn inhibited the malignant evolution of the tumor while restoring the viability and function of tumor-infiltrating immune cells.
Collapse
Affiliation(s)
- Boyu Su
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Qinjun Chen
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Xuwen Li
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Mingzhu Fang
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Yu Wang
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Haolin Song
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Haoyu You
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Zheng Zhou
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Yuxing Wu
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Zhenhao Zhao
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Yun Chen
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Hongrui Fan
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Chufeng Li
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Chen Jiang
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
- Department of Digestive DiseasesNational Regional Medical CenterBinhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350212China
| | - Tao Sun
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
- Quzhou Fudan InstituteQuzhou324003China
| |
Collapse
|
11
|
Qin ZX, Zuo L, Zeng Z, Ma R, Xie W, Zhu X, Zhou X. GalNac-siRNA conjugate delivery technology promotes the treatment of typical chronic liver diseases. Expert Opin Drug Deliv 2025; 22:455-469. [PMID: 39939158 DOI: 10.1080/17425247.2025.2466767] [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: 11/18/2024] [Revised: 01/26/2025] [Accepted: 02/10/2025] [Indexed: 02/14/2025]
Abstract
INTRODUCTION Nucleic acid-based therapeutics have become a key pillar of the 'third wave' of modern medicine, following the eras of small molecule inhibitors and antibody drugs. Their rapid progress is heavily dependent on delivery technologies, with the development of N-acetylgalactosamine (GalNAc) conjugates marking a breakthrough in targeting liver diseases. This technology has gained significant attention for its role in addressing chronic conditions like chronic hepatitis B (CHB) and nonalcoholic steatohepatitis (NASH), which are challenging to treat with conventional methods. AREAS COVERED This review explores the origins, mechanisms, and advantages of GalNAc-siRNA delivery systems, highlighting their ability to target hepatocytes via the asialoglycoprotein receptor (ASGPR). The literature reviewed covers preclinical and clinical advancements, particularly in CHB and NASH. Key developments in stabilization chemistry and conjugation technologies are examined, emphasizing their impact on enhancing therapeutic efficacy and patient compliance. EXPERT OPINION GalNAc-siRNA technology represents a transformative advancement in RNA interference (RNAi) therapies, addressing unmet needs in liver-targeted diseases. While significant progress has been made, challenges remain, including restricted targeting scope and scalability concerns. Continued innovation is expected to expand applications, improve delivery efficiency, and overcome limitations, establishing GalNAc-siRNA as a cornerstone for future nucleic acid-based treatments.
Collapse
Affiliation(s)
- Zhen-Xin Qin
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Ling Zuo
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Ziran Zeng
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Rongguan Ma
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Wenyan Xie
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
12
|
Zhang Q, Leng X, Peng L, Lin H, Xuan G, Zhang W, Mitomo H, Ijiro K, Wang G. Streamlining Bacterial Gene Regulation via Nucleic Acid Delivery with Gold Nanoclusters. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411723. [PMID: 39989200 DOI: 10.1002/smll.202411723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/06/2025] [Indexed: 02/25/2025]
Abstract
Delivery of exogenous nucleic acids (NAs) for gene regulation in bacteria, bypassing the barrier of the cell wall, is essential for advancing fundamental microbiology and genetic engineering, and the treatment of bacterial diseases. However, current methods that rely on electrical or chemical interventions are limited by their complexity, specialized expertise, and laboratory-specific instrumentation. This study explores the capability of gold nanoclusters (AuNCs) as carriers for delivering small-interfering RNA and antisense oligonucleotides into bacteria for targeted gene regulation while shielding them from degradation during transport. By enhancing the cytoplasmic membrane permeability, the AuNCs enable efficient internalization of NAs into both Gram-positive and Gram-negative bacteria while exerting negligible influence on bacterial activity. It is demonstrated that the rationally designed NAs can be released from the AuNCs within bacteria, enabling ~70% knockdown of mecA in Methicillin-resistant Staphylococcus aureus (MRSA). This significantly reduces MRSA's antibiotic resistance and enhances oxacillin treatment efficacy. Furthermore, the successful silencing of ligA in Escherichia coli and pilQ in Pseudomonas aeruginosa highlights the broad adaptability of the approach across diverse bacterial species. The AuNCs-based next-generation NA delivery system has the potential to transform bacterial gene regulation-previously restricted to laboratory settings-into a versatile and scalable solution for real-world application.
Collapse
Affiliation(s)
- Qingsong Zhang
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
| | - Xinyi Leng
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
| | - Lin Peng
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
| | - Hong Lin
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
| | - Guanhua Xuan
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, 169 Qixingnan Road, Ningbo, 315832, China
| | - Hideyuki Mitomo
- Research Institute for Electronic Science, Hokkaido University, Sapporo, 001-0021, Japan
| | - Kuniharu Ijiro
- Research Institute for Electronic Science, Hokkaido University, Sapporo, 001-0021, Japan
| | - Guoqing Wang
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao, 266404, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Centre, Qingdao, 266237, China
| |
Collapse
|
13
|
Wang M, Wang Y, Zhang P, Gu C, Zhao X, Gong X, Yang X, Pan J, Xi Y. Neutrophil-like cell membrane-coated metal-organic frameworks for siRNA delivery targeting NOX4 to alleviate oxidative stress in acute ischemic injury. Acta Biomater 2025; 196:487-505. [PMID: 40024424 DOI: 10.1016/j.actbio.2025.02.061] [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: 11/30/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Although reperfusion is the most effective treatment for acute ischemic stroke, it often results in serious secondary ischemia/reperfusion (I/R) injury due to oxidative stress. This oxidative stress primarily results from the overproduction of reactive oxygen species (ROS) during reperfusion which, in turn, is largely induced by high expression of NADPH oxidase 4 (NOX4). Inhibiting NOX4 gene expression has therefore been proposed as a direct approach to reduce ROS production and promote angiogenesis. Recognizing both the potential of siRNA-based therapies for selective gene silencing and the critical role of neutrophil-endothelial interactions during I/R injury, here we present a unique therapeutic approach where neutrophil-like cell membrane coated porous metal-organic framework nanoparticles are loaded with siNOX4 (M-MOF-siNOX4) and designed to target damaged brain microvascular tissue. These then mitigate oxidative stress by suppressing NOX4 expression. Using an in vitro oxygen-glucose deprivation/re-oxygenation model, we demonstrate that M-MOF-siNOX4 nanoparticles specifically bind to activated endothelial cells, effectively reducing NOX4 expression, decreasing both ROS production and cell apoptosis, and restoring cell viability. Use of an in vivo mouse model of middle cerebral artery occlusion further confirmed M-MOF-siNOX4 nanoparticles to substantially alleviate brain damage and protect neurological function following ischemic stroke. Taken together, our study presents an innovative and effective siRNA-based strategy for reducing oxidative stress in ischemic stroke therapy. STATEMENT OF SIGNIFICANCE: Ischemia/reperfusion (I/R) injury, a major complication of acute ischemic stroke, is primarily driven by oxidative stress due to the excessive production of reactive oxygen species (ROS). Current treatments targeting oxidative stress and cell death often lack specificity, leading to off-target effects. This study introduces an innovative nanoparticle-based therapy using neutrophil-like cell membrane-coated metal-organic frameworks (MOFs) to deliver siNOX4, an siRNA targeting NOX4, a key ROS-producing enzyme. This approach enhances targeted delivery, reduces ROS production and cell death, and significantly improves neurological recovery in stroke models. By overcoming the limitations of existing therapies, this strategy holds strong potential for revolutionizing ischemic stroke treatment and addressing other disorders related to oxidative stress.
Collapse
Affiliation(s)
- Min Wang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Yunbo Wang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Pengqi Zhang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Chenjie Gu
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Xianlei Zhao
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Xinghan Gong
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Xiaohang Yang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China.
| | - Jianwei Pan
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Yongmei Xi
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
14
|
Wang J, Huang H, Jia M, Chen S, Wang F, He G, Wu C, Lou K, Zheng X, Zhang H, Qin C, Yuan Y, Zen K, Liang H. Autologous platelet delivery of siRNAs by autologous plasma protein self-assembled nanoparticles for the treatment of acute kidney injury. J Nanobiotechnology 2025; 23:256. [PMID: 40156015 PMCID: PMC11954310 DOI: 10.1186/s12951-025-03338-6] [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: 10/05/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Acute kidney injury (AKI) involves the activation of intrarenal hemostatic and inflammatory pathways. Platelets rapidly migrate to affected sites of AKI and release extracellular vesicles (EVs) laden with bioactive mediators that regulate inflammation and hemostasis. While small interfering RNA (siRNA) is a potent gene-silencing tool for biomedical applications, its therapeutic application in vivo remains challenging. We developed an innovative nucleic acid delivery platform by hybridizing synthetic transformation-related protein 53 (p53) siRNA with autologous plasma and incubating the complex with autologous platelets. These engineered platelets selectively delivered p53 siRNA to injured renal tubular cells via EV-mediated cargo release, resulting in targeted p53 suppression in renal cells and subsequent attenuation of AKI progression. This platelet-centric translational strategy demonstrates significant potential for advancing precision therapies in AKI by exploiting endogenous platelet trafficking to deliver therapeutics directly to injury sites.
Collapse
Affiliation(s)
- Jiafan Wang
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Hai Huang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Meng Jia
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Si Chen
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fengjuan Wang
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Guiyang He
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Chong Wu
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Kaibin Lou
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Xuexue Zheng
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Heng Zhang
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Chao Qin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Yanggang Yuan
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Ke Zen
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China.
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, China.
| | - Hongwei Liang
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
15
|
Kim YM, Kim H, Park SC, Lee M, Jang MK. Targeted drug delivery of cancer cell-derived extracellular vesicles decorated with a VEGFR-binding peptide. Colloids Surf B Biointerfaces 2025; 252:114661. [PMID: 40203507 DOI: 10.1016/j.colsurfb.2025.114661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
Chemotherapy is commonly used to manage cancer lesions, but its use is limited or has a high risk of failure due to the side effects of the drugs. The use of a drug delivery system can minimize the side effects of drugs and maximize their anticancer effects. This study investigated the potential of tumor cell-derived small extracellular vesicles (sEV) as drug delivery vehicles for doxorubicin (Dox). In addition, the decoration of vascular endothelial growth factor receptor (VEGFR)-targeting peptide on the sEV provided an enhance specific cancer cell-targeting effect in vitro or homing capacity in vivo. The extrusion method was effective in loading Dox and displaying targeting peptide. The effective Dox release was resulted under acidic condition, an endosome pH. The growth of tumor masses on MCF-7 xenografted mice were significantly inhibited by this drug delivery system. We believe that our results will provide useful information for the development of chemotherapeutic drug delivery systems.
Collapse
Affiliation(s)
- Young-Min Kim
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea
| | - Hyeonseok Kim
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea
| | - Seong-Cheol Park
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea
| | - Mina Lee
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea.
| | - Mi-Kyeong Jang
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea.
| |
Collapse
|
16
|
Xiong S, Sun M, Zhang Y, Kong PR, Gan L, Gao L, Xu K, Wu HY, Zhu DY, Lin YH, Li R, Luo CX. Astrocytic BEST1 can serve as a target for functional recovery after ischemic stroke. Mol Ther 2025:S1525-0016(25)00196-0. [PMID: 40119514 DOI: 10.1016/j.ymthe.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/23/2024] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
Solid evidence from animal experiments supported the concept of peri-infarct tonic inhibition. Related drug targets have the potential to be translated for clinical stroke treatment. Recently, we reported the contribution of neuronal bestrophin-1 (BEST1)-mediated glutamate release to acute ischemic damage exacerbation in rodents. Now, we found a switch of abnormal BEST1 expression and function from neurons to astrocytes in the peri-infarct cortex following astrocytic activation. Excessive GABA was released through astrocytic BEST1 channel during the subacute phase of stroke, leading to sustained tonic inhibition. Astrocyte-specific knockdown of BEST1 promoted motor functional recovery, depending on reduced tonic inhibition. Moreover, we prepared self-assembled nanoparticles encapsulating siBest1 (SNP-siBest1), which displayed high brain accumulation and long circulation and knocked down astrocytic BEST1 effectively and safely. Systemic treatment with SNP-siBest1 after ischemic stroke showed a therapeutic effect in mice. Therefore, BEST1 is a potential target for stroke therapy from acute to subacute phase, and selective BEST1 blockers beyond nanoparticles are worth developing.
Collapse
Affiliation(s)
- Shuai Xiong
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Meng Sun
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ying Zhang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Pei-Ran Kong
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lu Gan
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ling Gao
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ke Xu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hai-Yin Wu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Dong-Ya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yu-Hui Lin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Rui Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Chun-Xia Luo
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
17
|
Laturski AE, Dulay MT, Perry JL, DeSimone JM. Transfection via RNA-Based Nanoparticles: Comparing Encapsulation vs Adsorption Approaches of RNA Incorporation. Bioconjug Chem 2025; 36:367-376. [PMID: 39999074 DOI: 10.1021/acs.bioconjchem.5c00028] [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: 02/27/2025]
Abstract
Historically, RNA delivery via nanoparticles has primarily relied on encapsulation, as demonstrated by lipid nanoparticles in SARS-CoV-2 vaccines. Concerns about RNA degradation on nanoparticle surfaces initially limited the exploration of adsorption-based approaches. However, recent advancements have renewed interest in adsorption as a viable alternative. This Viewpoint explores the approaches of RNA incorporation in nanoparticles, comparing encapsulation, adsorption, and the combination of encapsulation and adsorption, and presents a framework to guide the selection of the most suitable strategy based on general characteristics.
Collapse
Affiliation(s)
- Amy E Laturski
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Maria T Dulay
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Jillian L Perry
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7575, United States
| | - Joseph M DeSimone
- Department of Chemical Engineering and Department of Radiology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
18
|
Liu M, Wang Y, Zhang Y, Hu D, Tang L, Zhou B, Yang L. Landscape of small nucleic acid therapeutics: moving from the bench to the clinic as next-generation medicines. Signal Transduct Target Ther 2025; 10:73. [PMID: 40059188 PMCID: PMC11891339 DOI: 10.1038/s41392-024-02112-8] [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: 07/17/2024] [Revised: 10/23/2024] [Accepted: 12/13/2024] [Indexed: 03/17/2025] Open
Abstract
The ability of small nucleic acids to modulate gene expression via a range of processes has been widely explored. Compared with conventional treatments, small nucleic acid therapeutics have the potential to achieve long-lasting or even curative effects via gene editing. As a result of recent technological advances, efficient small nucleic acid delivery for therapeutic and biomedical applications has been achieved, accelerating their clinical translation. Here, we review the increasing number of small nucleic acid therapeutic classes and the most common chemical modifications and delivery platforms. We also discuss the key advances in the design, development and therapeutic application of each delivery platform. Furthermore, this review presents comprehensive profiles of currently approved small nucleic acid drugs, including 11 antisense oligonucleotides (ASOs), 2 aptamers and 6 siRNA drugs, summarizing their modifications, disease-specific mechanisms of action and delivery strategies. Other candidates whose clinical trial status has been recorded and updated are also discussed. We also consider strategic issues such as important safety considerations, novel vectors and hurdles for translating academic breakthroughs to the clinic. Small nucleic acid therapeutics have produced favorable results in clinical trials and have the potential to address previously "undruggable" targets, suggesting that they could be useful for guiding the development of additional clinical candidates.
Collapse
Affiliation(s)
- Mohan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yibing Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Die Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bailing Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
19
|
Xiao X, Yang S, Jiang G, He S. Current views and trends of nanomaterials as vectors for gene delivery since the 21st century: a bibliometric analysis. Nanomedicine (Lond) 2025; 20:439-454. [PMID: 39878523 PMCID: PMC11875476 DOI: 10.1080/17435889.2025.2457781] [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: 08/22/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Gene therapy is garnering increasing support due to its potential for a "once-delivered, lifelong benefit." The limitations of traditional gene delivery methods have spurred the advancement of bionanomaterials. Despite this progress, a thorough analysis of the evolution, current state, key contributors, focal studies, and future directions of nanomaterials in gene delivery remains absent. METHODS This study scrutinizes articles from the Web of Science, spanning 1 January 2 000, to 31 December 2023, employing various online tools for analysis and visualization. RESULTS The 21st century has witnessed consistent growth in scholarly work in this domain globally, with notable contributions from China and the US. At the same time, the Chinese Academy of Sciences (CAS), Harvard University, and Massachusetts Institute of Technology (MIT) have emerged as the most productive institutions, with CAS's academician Weihong Tan becoming the field's leading author. While drug delivery and nanoparticles (NPs) have been central themes for two decades, the research focus has shifted from modifying NPs and ultrafine particles to exploring polymer-hybrid NPs, mRNA vaccines, immune responses, green synthesis, and CRISPR/Cas tools. CONCLUSIONS This shift marks the transition from nanomaterials to bionanomaterials. The insights provided by this research offer a comprehensive overview of the field and valuable guidance for future investigations.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Sheng Yang
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ge Jiang
- Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shisheng He
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Zhao R, Guo J, Liu Z, Zhang Y, Zuo J, Lv S, Li X, Yao W, Zhang X. Zwitterionic lipid nanoparticles for efficient siRNA delivery and hypercholesterolemia therapy with rational charge self-transformation. Theranostics 2025; 15:3693-3712. [PMID: 40093884 PMCID: PMC11905131 DOI: 10.7150/thno.111685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Rationale: Effective delivery of small interfering RNA (siRNA) remains a significant challenge in treating hypercholesterolemia due to biocompatibility, cellular uptake, and endosomal escape issues. Rational regulation of carrier surface charge contributes to efficient siRNA delivery in vivo. Methods: This study introduces zwitterionic lipid nanoparticles (ZwiLNPs) as a novel solution to these challenges. By leveraging the unique properties of zwitterionic polymers, we achieved robust siRNA encapsulation and targeted delivery. The design of ZwiLNPs facilitates charge self-transformation in response to physiological conditions, which enhances their biocompatibility and cellular uptake efficiency. Result: In vivo studies demonstrated significant liver-targeting capabilities of ZwiLNPs, with improved endosomal escape following cellular internalization. Comparative analyses confirmed that ZwiLNPs outperform conventional lipid nanoparticles in terms of both cellular uptake and endosomal release. Conclusion: These findings position ZwiLNPs as a promising platform for RNA interference therapies, particularly for hypercholesterolemia and other lipid-related disorders.
Collapse
Affiliation(s)
- Ruichen Zhao
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Jing Guo
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Ziqi Liu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Yusheng Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Jiamin Zuo
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Songzhang Lv
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Wenlong Yao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing, 100191, PR China
| | - Xin Zhang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
21
|
Jin Y, Zhang B, Li J, Guo Z, Zhang C, Chen X, Ma L, Wang Z, Yang H, Li Y, Weng Y, Huang Y, Yan X, Fan K. Bioengineered protein nanocarrier facilitating siRNA escape from lysosomes for targeted RNAi therapy in glioblastoma. SCIENCE ADVANCES 2025; 11:eadr9266. [PMID: 39970222 PMCID: PMC11838010 DOI: 10.1126/sciadv.adr9266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025]
Abstract
RNA interference (RNAi) represents a promising gene-specific therapy against tumors. However, its clinical translation is impeded by poor performance of lysosomal escape and tumor targeting. This challenge is especially prominent in glioblastoma (GBM) therapy, necessitating the penetration of the blood-brain barrier (BBB). Leveraging the intrinsic tumor-targeting and BBB traversing capability of human H-ferritin, we designed a series of ferritin variants with positively charged cavity and truncated carboxyl terminus, termed tHFn(+). These nanocarriers respond to weak acid and disassemble in endosomal compartments, exposing the internal positive charges to facilitate the lysosomal escape of loaded small interfering RNA (siRNA). Functioning as universal siRNA nanocarriers, tHFn(+) significantly enhanced the uptake of different siRNAs and suppressed gene expressions associated with GBM progression. Furthermore, tHFn(+) traversed the BBB and targeted glioma in vivo by binding to its receptors (e.g., transferrin receptor 1). tHFn(+)-delivered siRNAs exhibited exceptional therapeutic effects against glioma in vivo, advancing RNAi therapeutics beyond GBM for the treatment of various diseases.
Collapse
Affiliation(s)
- Yiliang Jin
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Baoli Zhang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jianru Li
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenxi Guo
- Cryo-EM platform, School of Life Sciences, Peking University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Chen Zhang
- Cryo-EM platform, School of Life Sciences, Peking University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Xuehui Chen
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Long Ma
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhuoran Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Haiyin Yang
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; School of Medical Technology; Key Laboratory of Molecular Medicine and Biotherapy; Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Yong Li
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; School of Medical Technology; Key Laboratory of Molecular Medicine and Biotherapy; Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Yuhua Weng
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; School of Medical Technology; Key Laboratory of Molecular Medicine and Biotherapy; Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Yuanyu Huang
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; School of Medical Technology; Key Laboratory of Molecular Medicine and Biotherapy; Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering; Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan 451163, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan 451163, China
| |
Collapse
|
22
|
Márton É, Varga A, Domoszlai D, Buglyó G, Balázs A, Penyige A, Balogh I, Nagy B, Szilágyi M. Non-Coding RNAs in Cancer: Structure, Function, and Clinical Application. Cancers (Basel) 2025; 17:579. [PMID: 40002172 PMCID: PMC11853212 DOI: 10.3390/cancers17040579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
We are on the brink of a paradigm shift in both theoretical and clinical oncology. Genomic and transcriptomic profiling, alongside personalized approaches that account for individual patient variability, are increasingly shaping discourse. Discussions on the future of personalized cancer medicine are mainly dominated by the potential of non-coding RNAs (ncRNAs), which play a prominent role in cancer progression and metastasis formation by regulating the expression of oncogenic or tumor suppressor proteins at transcriptional and post-transcriptional levels; furthermore, their cell-free counterparts might be involved in intercellular communication. Non-coding RNAs are considered to be promising biomarker candidates for early diagnosis of cancer as well as potential therapeutic agents. This review aims to provide clarity amidst the vast body of literature by focusing on diverse species of ncRNAs, exploring the structure, origin, function, and potential clinical applications of miRNAs, siRNAs, lncRNAs, circRNAs, snRNAs, snoRNAs, eRNAs, paRNAs, YRNAs, vtRNAs, and piRNAs. We discuss molecular methods used for their detection or functional studies both in vitro and in vivo. We also address the challenges that must be overcome to enter a new era of cancer diagnosis and therapy that will reshape the future of oncology.
Collapse
Affiliation(s)
- Éva Márton
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - Alexandra Varga
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - Dóra Domoszlai
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - Gergely Buglyó
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - Anita Balázs
- Department of Integrative Health Sciences, Institute of Health Sciences, Faculty of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary;
| | - András Penyige
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - István Balogh
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Bálint Nagy
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| | - Melinda Szilágyi
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.M.); (A.V.); (D.D.); (G.B.); (A.P.); (I.B.); (B.N.)
| |
Collapse
|
23
|
Welden JR, Margvelani G, Miaro M, Mathews D, Rodgers DW, Stamm S. An oligo walk to identify siRNAs against the circular Tau 12->7 RNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635119. [PMID: 39974901 PMCID: PMC11838314 DOI: 10.1101/2025.01.27.635119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Circular RNAs are associated with numerous diseases and recent evidence shows that they can be translated into proteins after undergoing RNA modification. Circular RNAs differ from their 'linear' mRNA counterparts in their backsplice site, allowing selective targeting using RNA interference, which however limits the options to place the siRNA. We tested all possible siRNAs against the backsplice site of the circTau 12->7 RNA after it was subjected to adenosine to inosine RNA editing, a modification that promotes translation of the circRNA. Most siRNAs reduced the circRNA and protein abundance, which however did not correlate. We identified an siRNA with an IC50 of 750 pmol efficacy on protein expression. This circRNA fulfilled six of the eight criteria for siRNAs targeting mRNAs. Thus, modified circRNAs expressing protein can be targeted with siRNAs, but their optimal sequence needs to be determined empirically.
Collapse
|
24
|
Narasipura EA, Ma Y, Tiwade PB, VanKeulen-Miller R, Fung V, Fenton OS. A Chemoinformatic-Guided Synthesis of a Spleen-Expressing mRNA Lipid Nanoparticle Platform. Bioconjug Chem 2025; 36:54-65. [PMID: 39704424 DOI: 10.1021/acs.bioconjchem.4c00419] [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: 12/21/2024]
Abstract
mRNA lipid nanoparticles (LNPs) are a powerful technology that are actively being investigated for their ability to prevent, treat, and study disease. However, a major limitation remains: achieving extrahepatic mRNA expression. The development of new carriers could enable the expression of mRNA in non-liver targets, thus expanding the utility of mRNA-based medicines. In this study, we use a combination of chemoinformatic-guided material synthesis and design of experiment optimization for the development of a spleen-expressing lipid nanoparticle (SE-LNP). We begin with the synthesis of a novel cholesterol derivative followed by SE-LNP formulation and design of experiment-guided optimization to identify three lead SE-LNPs. We then evaluate their in vitro delivery mechanism, in vivo biodistribution, and protein expression in mice, ultimately achieving spleen-preferential expression. The goal of this paper is thus to create LNPs that preferentially express mRNA in the spleen upon intravenous delivery, demonstrating the potential of LNPs to modulate gene expression in extrahepatic tissues for disease treatment.
Collapse
Affiliation(s)
- Eshan A Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Palas Balakdas Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Vincent Fung
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
25
|
Kim M, Park S, Kim S, Seo J, Roh S. A Novel Cell-Penetrating Peptide-Vascular Endothelial Growth Factor Small Interfering Ribonucleic Acid Complex That Mediates the Inhibition of Angiogenesis by Human Umbilical Vein Endothelial Cells and in an Ex Vivo Mouse Aorta Ring Model. Biomater Res 2025; 29:0120. [PMID: 39780959 PMCID: PMC11704089 DOI: 10.34133/bmr.0120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Angiogenesis is mediated by vascular endothelial growth factor (VEGF), a protein that plays a key role in wound healing, inflammatory diseases, cardiovascular processes, ocular diseases, and tumor growth. Indeed, modulation of angiogenesis represents a potential approach to treating cancer and, as such, therapeutic approaches targeting VEGF and its receptors have been widely investigated as part of the broader search for curative interventions. Equally, RNA interference is a powerful tool for treating diseases, but its application as a disease treatment has been limited in part because of a lack of efficient small interfering RNA (siRNA) delivery systems. The purpose of this study was to characterize an amphipathic cell-penetrating peptide, Ara27, and its potential as an effective delivery vehicle as a conjugate with VEGF siRNA (siVEGF). In our study, we demonstrate that exposure of human umbilical vein endothelial cells (HUVECs) with Ara27-siVEGF complexes did not lead to cytotoxicity and can lead to down-regulation of cellular levels of both VEGF mRNA and protein. Moreover, treatment with the Ara27-siVEGF complex attenuates the phosphorylation of VEGFR2, Akt, and ERK in HUVECs and inhibits their capacity for wound healing and tube formation, both of which characteristics reflective of angiogenesis. In addition, we performed an ex vivo study to find that treatment with the Ara27-siVEGF complex inhibits aorta ring sprouting. Furthermore, the complex did not induce immunotoxicity in THP-1 and RAW264.7 cells. Taken together, our studies demonstrate that an Ara27-siVEGF conjugate is efficient for knockdown of VEGF in HUVECs to inhibit angiogenesis, without marked cytotoxic and immunotoxic effects.
Collapse
Affiliation(s)
- Minseo Kim
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute,
Seoul National University School of Dentistry, Seoul 08826, Republic of Korea
| | - Sangkyu Park
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute,
Seoul National University School of Dentistry, Seoul 08826, Republic of Korea
- Biomedical Research Institute,
NeoRegen Biotech Co., Ltd., Suwon, Gyeonggi 16614, Republic of Korea
| | - Soyi Kim
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute,
Seoul National University School of Dentistry, Seoul 08826, Republic of Korea
| | - Jeongmin Seo
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute,
Seoul National University School of Dentistry, Seoul 08826, Republic of Korea
- Biomedical Research Institute,
NeoRegen Biotech Co., Ltd., Suwon, Gyeonggi 16614, Republic of Korea
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute,
Seoul National University School of Dentistry, Seoul 08826, Republic of Korea
| |
Collapse
|
26
|
Dias MF, Cruz-Cazarim ELC, Pittella F, Baião A, Pacheco AC, Sarmento B, Fialho SL. Co-delivery of antioxidants and siRNA-VEGF: promising treatment for age-related macular degeneration. Drug Deliv Transl Res 2025:10.1007/s13346-024-01772-x. [PMID: 39751765 DOI: 10.1007/s13346-024-01772-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
Current treatments for retinal disorders are anti-angiogenic agents, laser photocoagulation, and photodynamic therapies. These conventional treatments focus on reducing abnormal blood vessel formation in the retina, which, in a low-oxygen environment, can lead to harmful proliferation of endothelial cells. This results in dysfunctional, leaky blood vessels that cause retinal edema, hemorrhage, and vision loss. Age-related Macular Degeneration is a primary cause of vision loss and blindness in the elderly, impacting around 20% of those over 50 years old. This complex disease is also closely related to oxidative stress in retina. In this review, we explore the challenge of treating retinal diseases, alternatives and possibilities of enhancing the effectiveness of therapies using co-delivery systems containing both antiangiogenic and antioxidant therapeutic agents. Despite recent proposals potential, the lack of extensive clinical studies on the long-term outcomes and optimal combinations of therapies means that the full risk profile and effectiveness of combined therapy are not yet completely understood. These factors must be carefully considered and managed by healthcare providers to optimize treatment outcomes and ensure patient safety.
Collapse
Affiliation(s)
- Marina F Dias
- Pharmaceutical Research and Development, Ezequiel Dias Foundation, Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, CEP 30510-010, Minas Gerais, Brazil
| | - Estael L C Cruz-Cazarim
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Juiz de Fora, Juiz de Fora, CEP 36036-900, Minas Gerais, Brazil
| | - Frederico Pittella
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Juiz de Fora, Juiz de Fora, CEP 36036-900, Minas Gerais, Brazil
| | - Ana Baião
- i3S - Instituto Nacional de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Engenharia Biomédica, INEB, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, ICBAS, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Ana Catarina Pacheco
- i3S - Instituto Nacional de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Engenharia Biomédica, INEB, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- CESPU-IUCS, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| | - Bruno Sarmento
- i3S - Instituto Nacional de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Engenharia Biomédica, INEB, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- CESPU-IUCS, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| | - Silvia L Fialho
- Pharmaceutical Research and Development, Ezequiel Dias Foundation, Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, CEP 30510-010, Minas Gerais, Brazil.
| |
Collapse
|
27
|
Hu D, Li Y, Yuan W, Ye X, Wang K, Xiao Y, Peng Z, Wu Q, Zeng C, Liu J, Zhou L. Bioactive cationic polymer-based hydrogel with Engrailed-1 gene silencing and microenvironment modulation for enhanced scarless diabetic wound healing. CHEMICAL ENGINEERING JOURNAL 2025; 504:158713. [DOI: 10.1016/j.cej.2024.158713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
28
|
Sindhi K, Kanugo A. Recent Developments in Nanotechnology and Immunotherapy for the Diagnosis and Treatment of Pancreatic Cancer. Curr Pharm Biotechnol 2025; 26:143-168. [PMID: 38415488 DOI: 10.2174/0113892010284407240212110745] [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: 10/31/2023] [Revised: 12/29/2023] [Accepted: 01/16/2024] [Indexed: 02/29/2024]
Abstract
Pancreatic cancer kills millions of people worldwide each year and is one of the most prevalent causes of mortality that requires prompt therapy. A large number of people suffering from pancreatic cancer are detected at an advanced stage, with incurable and drug-resistant tumor, hence the overall survival rate of pancreatic cancer is less. The advance phase of this cancer is generated because of expression of the cancer-causing gene, inactivation of the tumorsuppressing gene, and deregulation of molecules in different cellular signalling pathways. The prompt diagnosis through the biomarkers significantly evades the progress and accelerates the survival rates. The overexpression of Mesothelin, Urokinase plasminogen activator, IGFR, Epidermal growth factor receptor, Plectin-1, Mucin-1 and Zinc transporter 4 were recognized in the diagnosis of pancreatic cancer. Nanotechnology has led to the development of nanocarriersbased formulations (lipid, polymer, inorganic, carbon based and advanced nanocarriers) which overcome the hurdles of conventional therapy, chemotherapy and radiotherapy which causes toxicity to adjacent healthy tissues. The biocompatibility, toxicity and large-scale manufacturing are the hurdles associated with the nanocarriers-based approaches. Currently, Immunotherapybased techniques emerged as an efficient therapeutic alternative for the prevention of cancer. Immunological checkpoint targeting techniques have demonstrated significant efficacy in human cancers. Recent advancements in checkpoint inhibitors, adoptive T cell therapies, and cancer vaccines have shown potential in overcoming the immune evasion mechanisms of pancreatic cancer cells. Combining these immunotherapeutic approaches with nanocarriers holds great promise in enhancing the antitumor response and improving patient survival.
Collapse
Affiliation(s)
- Komal Sindhi
- Department of Pharmaceutics, SVKM NMIMS School of Pharmacy and Technology Management, Shirpur, 425405, India
| | - Abhishek Kanugo
- Department of Pharmaceutics, SVKM NMIMS School of Pharmacy and Technology Management, Shirpur, 425405, India
- Department of Pharmaceutical Quality Assurance, SVKM Institute of Pharmacy, Dhule, 424001, India
| |
Collapse
|
29
|
Zhang Y, Zhang M, Song H, Dai Q, Liu C. Tumor Microenvironment-Responsive Polymer-Based RNA Delivery Systems for Cancer Treatment. SMALL METHODS 2025; 9:e2400278. [PMID: 38803312 DOI: 10.1002/smtd.202400278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/30/2024] [Indexed: 05/29/2024]
Abstract
Ribonucleic acid (RNA) therapeutics offer a broad prospect in cancer treatment. However, their successful application requires overcoming various physiological barriers to effectively deliver RNAs to the target sites. Currently, a number of RNA delivery systems based on polymeric nanoparticles are developed to overcome these barriers in RNA delivery. This work provides an overview of the existing RNA therapeutics for cancer gene therapy, and particularly summarizes those that are entering the clinical phase. This work then discusses the core features and latest research developments of tumor microenvironment-responsive polymer-based RNA delivery carriers which are designed based on the pathological characteristics of the tumor microenvironment. Finally, this work also proposes opportunities for the transformation of RNA therapies into cancer immunotherapy methods in clinical applications.
Collapse
Affiliation(s)
- Yahan Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ming Zhang
- Department of Pathology, Peking University International Hospital, Beijing, 102206, China
| | - Haiqin Song
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Qiong Dai
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
30
|
Li S, Gao H, Wang H, Zhao X, Pan D, Pacheco-Fernández I, Ma M, Liu J, Hirvonen J, Liu Z, Santos HA. Tailored polysaccharide entrapping metal-organic framework for RNAi therapeutics and diagnostics in atherosclerosis. Bioact Mater 2025; 43:376-391. [PMID: 39399834 PMCID: PMC11470791 DOI: 10.1016/j.bioactmat.2024.08.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 10/15/2024] Open
Abstract
Metal-organic frameworks (MOFs) hold promise as theranostic carriers for atherosclerosis. However, to further advance their therapeutic effects with higher complexity and functionality, integrating multiple components with complex synthesis procedures are usually involved. Here, we reported a facile and general strategy to prepare multifunctional anti-atherosclerosis theranostic platform in a single-step manner. A custom-designed multifunctional polymer, poly(butyl methacrylate-co-methacrylic acid) branched phosphorylated β-glucan (PBMMA-PG), can effectively entrap different MOFs via coordination, simultaneously endow the MOF with enhanced stability, lesional macrophages selectivity and enhanced endosome escape. Sequential ex situ characterization and computational studies elaborated the potential mechanism. This facile post-synthetic modification granted the administered nanoparticles atherosclerotic tropism by targeting Dectin-1+ macrophages, enhancing in situ MR signal intensity by 72 %. Delivery of siNLRP3 effectively mitigated NLRP3 inflammasomes activation, resulting a 43 % reduction of plaque area. Overall, the current study highlights a simple and general approach for fabricating a MOF-based theranostic platform towards atherosclerosis conditioning, which may also expand to other indications targeting the lesional macrophages.
Collapse
Affiliation(s)
- Sen Li
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Han Gao
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Haoji Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Xiaolin Zhao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Da Pan
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, 210009, PR China
| | - Idaira Pacheco-Fernández
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Ming Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Jianjun Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Zehua Liu
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Hélder A. Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
31
|
Krishnaswami V, Janakiraman K, Sethuraman V, Raja J, Muruganantham S, Chelladurai S. Recent Insights into Nano-mediated siRNA Drug Delivery. Curr Drug Metab 2025; 25:554-563. [PMID: 39716803 DOI: 10.2174/0113892002339055241211050131] [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: 07/10/2024] [Revised: 11/05/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024]
Abstract
Gene silencing is the characteristic that inhibits gene expression afforded by siRNA interference. The efficacy of the delivery system in terms of precision, efficacy, and stability can be enhanced by genebased drug delivery options. The delivery challenges and their associated side effects create a challenge for the delivery of gene-based drug delivery carriers. Nano-based delivery systems were reported to improve the efficacy of therapy. The absence of an efficient delivery mechanism that shields siRNA from nuclease degradation delivers it to cancer cells, and releases it into the cytoplasm of specific cancer cells without causing side effects is currently the greatest obstacle to the practical implementation of siRNA therapy. This article focuses on general aspects of siRNA and various siRNA nanocarrier-based formulations. In the near future, we will move towards the siRNA-based drug delivery approach.
Collapse
Affiliation(s)
| | - Kumar Janakiraman
- Department of Biotechnology, Rathinam Technical Campus, Eachanari, Coimbatore, 641021, India
| | - Vaidevi Sethuraman
- Department of Biotechnology, Rathinam Technical Campus, Eachanari, Coimbatore, 641021, India
| | - Jacob Raja
- Department of Periodontology, Rajas Dental College and Hospital, Kavalkinaru, 627105, Tirunelveli, Tamil Nadu, India
| | - Selvakumar Muruganantham
- Department of Pharmaceutics, Vivekanandha Pharmacy College for Women, Sankari, Salem, 637303, Tamil Nadu, India
| | - Senthilkumar Chelladurai
- Department of Pharmaceutics, Sir Issac Newton College of Pharmacy, Pappakovil, Nagapatinam, Tamil Nadu, India
| |
Collapse
|
32
|
Wang H, Cheng Y. Polymers for mRNA Delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70002. [PMID: 39763235 DOI: 10.1002/wnan.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/22/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
mRNA delivery has emerged as a transformative approach in biotechnology and medicine, offering a versatile platform for the development of novel therapeutics. Unlike traditional small molecule drugs or protein-based biologics, mRNA therapeutics have the unique ability to direct cells to generate therapeutic proteins, allowing for precise modulation of biological processes. The delivery of mRNA into target cells is a critical step in realizing the therapeutic potential of this technology. In this review, our focus is on the latest advancements in designing functional polymers to achieve efficient mRNA delivery. Biodegradable polymers and low molecular weight polymers in addressing the balance in mRNA binding and release are summarized. Benefiting from the excellent performance of lipid nanoparticles in mRNA delivery, polymer/lipid hybrid nanostructures are also included. Finally, the challenges and future prospects in the development of polymer-based mRNA delivery systems are discussed.
Collapse
Affiliation(s)
- Hui Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, China
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
33
|
Askarizadeh A, Vahdat-Lasemi F, Karav S, Kesharwani P, Sahebkar A. Lipid nanoparticle-based delivery of small interfering RNAs: New possibilities in the treatment of diverse diseases. Eur Polym J 2025; 223:113624. [DOI: 10.1016/j.eurpolymj.2024.113624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
34
|
Beňačka R, Szabóová D, Guľašová Z, Hertelyová Z. Non-Coding RNAs in Breast Cancer: Diagnostic and Therapeutic Implications. Int J Mol Sci 2024; 26:127. [PMID: 39795985 PMCID: PMC11719911 DOI: 10.3390/ijms26010127] [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: 11/24/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Breast cancer (BC) is one of the most prevalent forms of cancer globally, and has recently become the leading cause of cancer-related mortality in women. BC is a heterogeneous disease comprising various histopathological and molecular subtypes with differing levels of malignancy, and each patient has an individual prognosis. Etiology and pathogenesis are complex and involve a considerable number of genetic alterations and dozens of alterations in non-coding RNA expression. Non-coding RNAs are part of an abundant family of single-stranded RNA molecules acting as key regulators in DNA replication, mRNA processing and translation, cell differentiation, growth, and overall genomic stability. In the context of breast cancer, non-coding RNAs are involved in cell cycle control and tumor cell migration and invasion, as well as treatment resistance. Alterations in non-coding RNA expression may contribute to the development and progression of breast cancer, making them promising biomarkers and targets for novel therapeutic approaches. Currently, the use of non-coding RNAs has not yet been applied to routine practice; however, their potential has been very well studied. The present review is a literature overview of current knowledge and its objective is to delineate the function of diverse classes of non-coding RNAs in breast cancer, with a particular emphasis on their potential utility as diagnostic and prognostic markers or as therapeutic targets and tools.
Collapse
Affiliation(s)
- Roman Beňačka
- Department of Pathophysiology, Medical Faculty, Pavol Jozef Šafarik University, 04011 Košice, Slovakia;
| | - Daniela Szabóová
- Department of Pathophysiology, Medical Faculty, Pavol Jozef Šafarik University, 04011 Košice, Slovakia;
| | - Zuzana Guľašová
- Center of Clinical and Preclinical Research MEDIPARK, Pavol Jozef Šafarik University, 04011 Košice, Slovakia; (Z.G.); (Z.H.)
| | - Zdenka Hertelyová
- Center of Clinical and Preclinical Research MEDIPARK, Pavol Jozef Šafarik University, 04011 Košice, Slovakia; (Z.G.); (Z.H.)
| |
Collapse
|
35
|
Wu S, Yang Y, Lian X, Zhang F, Hu C, Tsien J, Chen Z, Sun Y, Vaidya A, Kim M, Sung YC, Xiao Y, Bian X, Wang X, Tian Z, Guerrero E, Robinson J, Basak P, Qin T, Siegwart DJ. Isosteric 3D Bicyclo[1.1.1]Pentane (BCP) Core-Based Lipids for mRNA Delivery and CRISPR/Cas Gene Editing. J Am Chem Soc 2024; 146:34733-34742. [PMID: 39655603 PMCID: PMC11717372 DOI: 10.1021/jacs.4c13154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Lipid nanoparticles (LNPs) are an essential component of messenger RNA (mRNA) vaccines and genome editing therapeutics. Ionizable amino lipids, which play the most crucial role in enabling mRNA to overcome delivery barriers, have, to date, been restricted to two-dimensional (2D) architectures. Inspired by improved physicochemical properties resulting from the incorporation of three-dimensionality (3D) into small-molecule drugs, we report the creation of 3D ionizable lipid designs through the introduction of bicyclo[1.1.1]pentane (BCP) core motifs. BCP-based lipids enabled efficient in vivo mRNA delivery to the liver and spleen with significantly greater performance over 2D benzene- and cyclohexane-based analogues. Notably, lead BCP-NC2-C12 LNPs mediated ∼90% reduction in the PCSK9 serum protein level via CRISPR/Cas9 gene knockout, outperforming 2D controls and clinically used DLin-MC3-DMA LNPs at the same dose. Here, we introduce BCP-based designs with superior in vivo activity, thereby expanding the chemical scope of ionizable amino lipids from 2D to 3D and offering a promising avenue to improve mRNA and gene editing efficiency for the continued development of genetic medicines.
Collapse
Affiliation(s)
- Shiying Wu
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Yangyang Yang
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Xizhen Lian
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Fangyu Zhang
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Chao Hu
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Jet Tsien
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Zexiang Chen
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Yehui Sun
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Amogh Vaidya
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Minjeong Kim
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Yun-Chieh Sung
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Yufen Xiao
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Xiaoyan Bian
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Xu Wang
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Zeru Tian
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Erick Guerrero
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Joshua Robinson
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Pratima Basak
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Tian Qin
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Daniel J Siegwart
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| |
Collapse
|
36
|
Gusev AE, Ivanov VN, Dmitriev NA, Kholstov AV, Vasilichin VA, Kofiadi IA, Khaitov MR. A Novel Pot-Economy Approach to the Synthesis of Triantennary GalNAc-Oligonucleotide. Molecules 2024; 29:5959. [PMID: 39770048 PMCID: PMC11679575 DOI: 10.3390/molecules29245959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/30/2025] Open
Abstract
N-Acetylgalactosamine (GalNAc) is an efficient and multifunctional delivery tool in the development and synthesis of chemically modified oligonucleotide therapeutics (conjugates). Such therapeutics demonstrate improved potency in vivo due to the selective and efficient delivery to hepatocytes in the liver via receptor-mediated endocytosis, which is what drives the high interest in this molecule. The ways to synthesize such structures are relatively new and have not been optimized in terms of the yields and stages both in lab and large-scale synthesis. Another significant criterion, especially in large-scale synthesis, is to match ecological norms and perform the synthesis in accordance with the Green Chemistry approach, i.e., to control and minimize the amounts of reagents and resources consumed and the waste generated. Here, we provide a robust and resource effective pot-economy method for the synthesis of triantennary GalNAc and GalNAc phosphoramidite/CPG optimized for laboratory scales.
Collapse
|
37
|
Cheng J, Jian L, Chen Z, Li Z, Yu Y, Wu Y. In Vivo Delivery Processes and Development Strategies of Lipid Nanoparticles. Chembiochem 2024; 25:e202400481. [PMID: 39101874 DOI: 10.1002/cbic.202400481] [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: 05/31/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/06/2024]
Abstract
Lipid nanoparticles (LNPs) represent an advanced and highly efficient delivery system for RNA molecules, demonstrating exceptional biocompatibility and remarkable delivery efficiency. This is evidenced by the clinical authorization of three LNP formulations: Patisiran, BNT162b2, and mRNA-1273. To further maximize the efficacy of RNA-based therapy, it is imperative to develop more potent LNP delivery systems that can effectively protect inherently unstable and negatively charged RNA molecules from degradation by nucleases, while facilitating their cellular uptake into target cells. Therefore, this review presents feasible strategies commonly employed for the development of efficient LNP delivery systems. The strategies encompass combinatorial chemistry for large-scale synthesis of ionizable lipids, rational design strategy of ionizable lipids, functional molecules-derived lipid molecules, the optimization of LNP formulations, and the adjustment of particle size and charge property of LNPs. Prior to introducing these developing strategies, in vivo delivery processes of LNPs, a crucial determinant influencing the clinical translation of LNP formulations, is described to better understand how to develop LNP delivery systems.
Collapse
Affiliation(s)
- Jiashun Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lina Jian
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhaolin Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhuoyuan Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yaobang Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yihang Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
38
|
Lou W, Zhang L, Wang J. Current status of nucleic acid therapy and its new progress in cancer treatment. Int Immunopharmacol 2024; 142:113157. [PMID: 39288629 DOI: 10.1016/j.intimp.2024.113157] [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: 06/11/2024] [Revised: 07/05/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Nucleic acid is an essential biopolymer in all living cells, performing the functions of storing and transmitting genetic information and synthesizing protein. In recent decades, with the progress of science and biotechnology and the continuous exploration of the functions performed by nucleic acid, more and more studies have confirmed that nucleic acid therapy for living organisms has great medical therapeutic potential. Nucleic acid drugs began to become independent therapeutic agents. As a new therapeutic method, nucleic acid therapy plays an important role in the treatment of genetic diseases, viral infections and cancers. There are currently 19 nucleic acid drugs approved by the Food and Drug Administration (FDA). In the following review, we start from principles and advantages of nucleic acid therapy, and briefly describe development history of nucleic acid drugs. And then we give examples of various RNA therapeutic drugs, including antisense oligonucleotides (ASO), mRNA vaccines, small interfering RNA (siRNA) and microRNA (miRNA), aptamers, and small activating RNA (saRNA). In addition, we also focused on the current status of nucleic acid drugs used in cancer therapy and the breakthrough in recent years. Clinical trials of nucleic acid drugs for cancer treatment are under way, conventional radiotherapy and chemotherapy combined with the immunotherapies such as checkpoint inhibitors and nucleic acid drugs may be the main prospects for successful cancer treatment.
Collapse
Affiliation(s)
- Wenting Lou
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Leqi Zhang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Jianwei Wang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China; Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 2nd Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou 310009, China.
| |
Collapse
|
39
|
Li Y, Zhang J, Ma B, Yu W, Xu M, Luan W, Yu Q, Zhang L, Rong R, Fu Y, Cao H. Nanotechnology used for siRNA delivery for the treatment of neurodegenerative diseases: Focusing on Alzheimer's disease and Parkinson's disease. Int J Pharm 2024; 666:124786. [PMID: 39378955 DOI: 10.1016/j.ijpharm.2024.124786] [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: 06/19/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024]
Abstract
Neurodegenerative diseases (ND) are often accompanied by dementia, motor dysfunction, or disability. Caring for these patients imposes a significant psychological and financial burden on families. Until now, there are no effective methods for the treatment of NDs. Among them, Alzheimer's disease (AD) and Parkinson's disease (PD) are the two most common. Recently, studies have revealed that the overexpression of certain genes may be linked to the occurrence of AD and PD. Small interfering RNAs (siRNAs) are a powerful tool for gene silencing because they can specifically bind to and cleave target mRNA. However, the intrinsic properties of naked siRNA and various physiological barriers limit the application of siRNA in the brain. Nanotechnology is a promising option for addressing these issues. Nanoparticles are not only able to protect siRNA from degradation but also have the advantage of crossing various physiological barriers to reach the brain target of siRNA. In this review, we aim to introduce diverse nanotechnology used for delivering siRNA to treat AD and PD. Finally, we will briefly discuss our perspectives on this promising field.
Collapse
Affiliation(s)
- Yan Li
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Jiahui Zhang
- School of Pharmacy, Yantai University, Yantai 264005, China
| | - Boqin Ma
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Wenjun Yu
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Meixia Xu
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Weijing Luan
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Qinglong Yu
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Li Zhang
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Rong Rong
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China.
| | - Yuanlei Fu
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China.
| | - Haiqiang Cao
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
40
|
Uz M, Bulmus V, Alsoy Altinkaya S. Comparison of Cell-Penetrating and Fusogenic TAT-HA2 Peptide Performance in Peptideplex, Multicomponent, and Conjugate siRNA Delivery Systems. ACS OMEGA 2024; 9:47461-47474. [PMID: 39651078 PMCID: PMC11618417 DOI: 10.1021/acsomega.4c05808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/01/2024] [Accepted: 11/12/2024] [Indexed: 12/11/2024]
Abstract
In this study, the performance of the cell-penetrating and fusogenic peptide, TAT-HA2, which consists of a cell-permeable HIV trans-activator of transcription (TAT) protein transduction domain and a pH-responsive influenza A virus hemagglutinin protein (HA2) domain, was comparatively evaluated for the first time in peptideplex, multicomponent, and conjugate siRNA delivery systems. TAT-HA2 in all three systems protected siRNA from degradation, except in the conjugate system with a low Peptide/siRNA ratio. The synergistic effect of different peptide domains enhanced the transfection efficiency of multicomponent and conjugate systems compared to that of peptideplexes, which was attributed to the surface configuration of TAT-HA2 peptides depending on the nature of attachment. Particularly, the multicomponent system showed better cellular uptake and endosomal escape than the peptideplexes, resulting in enhanced siRNA delivery in the cytoplasm. In addition, the presence of cleavable disulfide bonds in multicomponent and conjugate systems promoted the effective siRNA delivery in the cytoplasm, resulting in improved gene silencing activity. The multicomponent system reduced the level of luciferase expression in SKOV3 cells to 45% (±4). In contrast, the conjugate system and the commercially available siRNA transfection agent, Lipofectamine RNAiMax, caused luciferase suppression down to 55% (±2) at a siRNA dose of 100 nM. For the same dose, the peptideplex system could only reduce the luciferase expression to 65% (±5). None of the developed systems showed significant toxicity at any dose. Overall, the TAT-HA2 peptide is promising as a siRNA delivery vector; however, its performance depends on the nature of attachment and, as a result, its surface configuration on the developed delivery system.
Collapse
Affiliation(s)
- Metin Uz
- Department of Chemical and Biomedical Engineering, Cleveland State University, 2121 Euclid Ave., FH 455, Cleveland, Ohio 44115-2214, United States
| | - Volga Bulmus
- Department of Bioengineering, Izmir Institute of Technology, Gulbahce Koyu/Urla, Izmir 35430, Turkey
| | - Sacide Alsoy Altinkaya
- Department of Chemical Engineering, Izmir Institute of Technology, Gulbahce Koyu/Urla, Izmir 35430, Turkey
| |
Collapse
|
41
|
Amar‐Lewis E, Cohen L, Chintakunta R, Benafsha C, Lavi Y, Goldbart R, Traitel T, Gheber LA, Kost J. Elucidating siRNA Cellular Delivery Mechanism Mediated by Quaternized Starch Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2405524. [PMID: 39359045 PMCID: PMC11657042 DOI: 10.1002/smll.202405524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/30/2024] [Indexed: 10/04/2024]
Abstract
Starch-based nanoparticles are highly utilized in the realm of drug delivery taking advantage of their biocompatibility and biodegradability. Studies have utilized Quaternized starch (Q-starch) for small interfering RNA (siRNA) delivery, in which quaternary amines enable interaction with negatively charged siRNA, resulting in self-assembly complexation. Although reports present numerous applications, the demonstrated efficacy is nonetheless limited due to undiscovered cellular mechanistic delivery. In this study, a deep dive into Q-starch/siRNA complexes' cellular mechanism and kinetics at the cellular level is revealed using single-particle tracking and cell population level using imaging flow cytometry. Uptake studies depict the efficient cellular internalization via endocytosis while a significant fraction of complexes' intracellular fate is lysosome. Utilizing single-particle tracking, it is found that an average of 15% of cellular detected complexes escape the endosome which holds the potential for the integration in the cytoplasmatic gene silencing mechanism. Additional experimental manipulations (overcoming endosomal escape) demonstrate that the complex's disassembly is the rate-limiting step, correlating Q-starch's structure-function properties as siRNA carrier. Structure-function properties accentuating the high affinity of the interaction between Q-starch's quaternary groups and siRNA's phosphate groups that results in low release efficiency. However, low-frequency ultrasound (20 kHz) application may have induced siRNA release resulting in faster gene silencing kinetics.
Collapse
Affiliation(s)
- Eliz Amar‐Lewis
- Ilse Katz Institute for Nanoscale Science & TechnologyBen‐Gurion University of the NegevBeer‐Sheva84105Israel
- Department of Chemical EngineeringBen‐Gurion University of the NegevBeer‐Sheva84105Israel
| | - Limor Cohen
- Department of Biotechnology EngineeringBen‐Gurion University of the NegevBeer‐Sheva84105Israel
| | - Ramesh Chintakunta
- Department of Chemical EngineeringBen‐Gurion University of the NegevBeer‐Sheva84105Israel
| | - Chen Benafsha
- Department of Chemical EngineeringBen‐Gurion University of the NegevBeer‐Sheva84105Israel
| | - Yael Lavi
- Department of Biotechnology EngineeringBen‐Gurion University of the NegevBeer‐Sheva84105Israel
| | - Riki Goldbart
- Department of Chemical EngineeringBen‐Gurion University of the NegevBeer‐Sheva84105Israel
| | - Tamar Traitel
- Department of Chemical EngineeringBen‐Gurion University of the NegevBeer‐Sheva84105Israel
| | - Levi A. Gheber
- Department of Biotechnology EngineeringBen‐Gurion University of the NegevBeer‐Sheva84105Israel
| | - Joseph Kost
- Ilse Katz Institute for Nanoscale Science & TechnologyBen‐Gurion University of the NegevBeer‐Sheva84105Israel
- Department of Chemical EngineeringBen‐Gurion University of the NegevBeer‐Sheva84105Israel
| |
Collapse
|
42
|
Mirhadi E, Kesharwani P, Jha SK, Karav S, Sahebkar A. Utilizing ionic liquids as eco-friendly and sustainable carriers for delivering nucleic acids: A review on the revolutionary advancement in nano delivery systems. Int J Biol Macromol 2024; 283:137582. [PMID: 39542300 DOI: 10.1016/j.ijbiomac.2024.137582] [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: 09/07/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Ionic liquids (ILs) are an extremely versatile class of chemicals. It has been shown that they can effectively pass through many biological barriers in the human body to deliver medications. ILs are solvents noted for their ecological friendliness; they contain equal amounts of cations and anions and remain liquid at temperatures below 100 °C. Hence, these are ideal for biomedical applications owing to their advantageous properties such as biocompatibility, solubility, and adaptability. ILs are widely reported to improve the solubility and stability of nucleic acids (DNA and RNA) in aqueous conditions, allowing for more effective delivery. Certain ILs have shown the ability to enhance the absorption of nucleic acids into cells. In addition, ILs can also be used to create vectors for gene delivery, such as liposomes and nanoparticles, thereby improving the transfection efficiency of plasmid DNA and siRNA. Subsequently, the application of ILs for nucleic acid delivery has increased significantly in recent years. In this context, we believe that using ILs to enhance the transport of nucleic acids will have a considerable effect as a novel and crucial therapeutic method in the upcoming decades. The use of ILs as solvents to preserve the natural structure of DNA and RNA shows promise for a variety of biotechnological and medical applications. Notably, ILs may be utilized for a variety of functions, including extracting, concentrating, stabilizing, and spreading nucleic acids inside cells. Our review emphasizes the key findings of research works published in this domain, wherein outstanding effectiveness of delivering RNA to the desired areas was achieved, and was made possible through the utilization of ILs.
Collapse
Affiliation(s)
- Elaheh Mirhadi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Saurav Kumar Jha
- Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur 208016, Uttar Pradesh, India
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
43
|
Zhang Y, Yang T, Yang Y, Xu D, Hu Y, Zhang S, Luo N, Ning L, Ren L. siRNAEfficacyDB: An experimentally supported small interfering RNA efficacy database. IET Syst Biol 2024; 18:199-207. [PMID: 39541343 DOI: 10.1049/syb2.12102] [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: 07/04/2024] [Revised: 09/26/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024] Open
Abstract
Small interfering RNA (siRNA) has revolutionised biomedical research and drug development through precise post-transcriptional gene silencing technology. Despite its immense potential, siRNA therapy still faces technical challenges, such as delivery efficiency, targeting specificity, and molecular stability. To address these challenges and facilitate siRNA drug development, we have developed siRNAEfficacyDB, a comprehensive database that integrates experimentally validated siRNA efficacy data. This database contains 3544 siRNA records, covering 42 target genes and 5 cell lines. It provides detailed information on siRNA sequences, target genes, inhibition efficiencies, experimental techniques, cell lines, siRNA concentrations, and incubation times. siRNAEfficacyDB offers a user-friendly web interface that makes it easy to query, browse and analyse data, enabling efficient access to siRNA-related information. In summary, siRNAEfficacyDB provides a useful data foundation for siRNA drug design and optimisation, serving as a valuable resource for advancing computer-aided siRNA design research and nucleic acid drug development. siRNAEfficacyDB is freely available at https://cellknowledge.com.cn/siRNAEfficacy for non-commercial use.
Collapse
Affiliation(s)
- Yang Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Yang
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| | - Yu Yang
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| | - Dongsheng Xu
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| | - Yucheng Hu
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| | - Shuo Zhang
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| | - Nanchao Luo
- School of Computer Science and Technology, Aba Teachers College, Aba, Sichuan, China
| | - Lin Ning
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| | - Liping Ren
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| |
Collapse
|
44
|
Qi Y, Wang C, Lang H, Wang Y, Wang X, Zheng H, Lu Y. Liposome-based RNAi delivery in honeybee for inhibiting parasite Nosema ceranae. Synth Syst Biotechnol 2024; 9:853-860. [PMID: 39139857 PMCID: PMC11320372 DOI: 10.1016/j.synbio.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/14/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
Nosema ceranae, a parasite that parasitizes and reproduces in the gut of honeybees, has become a serious threat to the global apiculture industry. RNA interference (RNAi) technology can be used to inhibit N. ceranae growth by targeting silencing the thioredoxin reductase (TrxR) in N. ceranae. However, suitable carriers are one of the reasons limiting the application of RNAi due to the easy degradation of dsRNA in honeybees. As a vesicle composed of a lipid bilayer, liposomes are a good carrier for nucleic acid delivery, but studies in honeybees are lacking. In this study, liposomes were used for double-stranded RNA (dsRNA) dsTrxR delivery triggering RNAi to inhibit the N. ceranae growth in honeybees. Compared to naked dsTrxR, liposome-dsTrxR reduced N. ceranae numbers in the midgut and partially restored midgut morphology without affecting bee survival and gut microbial composition. The results of this study confirmed that liposomes could effectively protect dsRNA from entering the honeybee gut and provide a reference for using RNAi technology to suppress honeybee pests and diseases.
Collapse
Affiliation(s)
- Yue Qi
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Chen Wang
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Haoyu Lang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yueyi Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Xiaofei Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Hao Zheng
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
45
|
Sun X, Setrerrahmane S, Li C, Hu J, Xu H. Nucleic acid drugs: recent progress and future perspectives. Signal Transduct Target Ther 2024; 9:316. [PMID: 39609384 PMCID: PMC11604671 DOI: 10.1038/s41392-024-02035-4] [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: 10/31/2023] [Revised: 09/20/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024] Open
Abstract
High efficacy, selectivity and cellular targeting of therapeutic agents has been an active area of investigation for decades. Currently, most clinically approved therapeutics are small molecules or protein/antibody biologics. Targeted action of small molecule drugs remains a challenge in medicine. In addition, many diseases are considered 'undruggable' using standard biomacromolecules. Many of these challenges however, can be addressed using nucleic therapeutics. Nucleic acid drugs (NADs) are a new generation of gene-editing modalities characterized by their high efficiency and rapid development, which have become an active research topic in new drug development field. However, many factors, including their low stability, short half-life, high immunogenicity, tissue targeting, cellular uptake, and endosomal escape, hamper the delivery and clinical application of NADs. Scientists have used chemical modification techniques to improve the physicochemical properties of NADs. In contrast, modified NADs typically require carriers to enter target cells and reach specific intracellular locations. Multiple delivery approaches have been developed to effectively improve intracellular delivery and the in vivo bioavailability of NADs. Several NADs have entered the clinical trial recently, and some have been approved for therapeutic use in different fields. This review summarizes NADs development and evolution and introduces NADs classifications and general delivery strategies, highlighting their success in clinical applications. Additionally, this review discusses the limitations and potential future applications of NADs as gene therapy candidates.
Collapse
Affiliation(s)
- Xiaoyi Sun
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | | | - Chencheng Li
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Jialiang Hu
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Hanmei Xu
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
46
|
Grisorio L, Bongianino R, Gianeselli M, Priori SG. Gene therapy for cardiac diseases: methods, challenges, and future directions. Cardiovasc Res 2024; 120:1664-1682. [PMID: 39302117 DOI: 10.1093/cvr/cvae207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 09/22/2024] Open
Abstract
Gene therapy is advancing at an unprecedented pace, and the recent success of clinical trials reinforces optimism and trust among the scientific community. Recently, the cardiac gene therapy pipeline, which had progressed more slowly than in other fields, has begun to advance, overcoming biological and technical challenges, particularly in treating genetic heart pathologies. The primary rationale behind the focus on monogenic cardiac diseases is the well-defined molecular mechanisms driving their phenotypes, directly linked to the pathogenicity of single genetic mutations. This aspect makes these conditions a remarkable example of 'genetically druggable' diseases. Unfortunately, current treatments for these life-threatening disorders are few and often poorly effective, underscoring the need to develop therapies to modulate or correct their molecular substrates. In this review we examine the latest advancements in cardiac gene therapy, discussing the pros and cons of different molecular approaches and delivery vectors, with a focus on their therapeutic application in cardiac inherited diseases. Additionally, we highlight the key factors that may enhance clinical translation, drawing insights from previous trials and the current prospects of cardiac gene therapy.
Collapse
Affiliation(s)
- Luca Grisorio
- Department of Internal Medicine, University of Pavia, Via Golgi 19, Pavia, 27100, Italy
| | - Rossana Bongianino
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Via Maugeri 10, Pavia, 27100, Italy
| | - Matteo Gianeselli
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA
| | - Silvia Giuliana Priori
- Department of Internal Medicine, University of Pavia, Via Golgi 19, Pavia, 27100, Italy
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Via Maugeri 10, Pavia, 27100, Italy
- Molecular Cardiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), C/ Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| |
Collapse
|
47
|
Xu J, Li R, Yan D, Zhu L. Biomimetic Modification of siRNA/Chemo Drug Nanoassemblies for Targeted Combination Therapy in Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59765-59776. [PMID: 39447113 DOI: 10.1021/acsami.4c11064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
The development and progression of tumors are characterized by intricate biological processes. Monotherapy not only struggles to achieve effective treatment but also tends to precipitate a series of issues, including multidrug resistance and limited antitumor effect. Consequently, it is imperative to adopt a synergistic multitherapy approach to enhance the efficacy of tumor treatment. The integration of chemotherapy drug with oligonucleotide drug for combinational treatment has shown significant promise in improving tumor therapeutic efficiency. However, the effective in vivo codelivery of oligonucleotide drugs and chemotherapy drugs faces substantial challenges such as poor stability of oligonucleotide drugs during the circulation time, limited tumor accumulation, and uncertain delivery ratios of different payloads. To overcome these obstacles, we have engineered cyclic Arg-Gly-Asp (cRGD)-modified red blood cell membrane (RBCm)-coated multidrug nanocomplexes, which were self-assembled from the Polo-like kinase 1 siRNA (siPlk1) and an irreversible tyrosine kinase inhibitor neratinib targeted to human epidermal growth factor receptor 2 (HER2) overexpressed in breast cancer. Through electrostatic and amphiphilic interactions between the positively charged neratinib and negatively charged siPlk1, we have successfully fabricated uniform multidrug nanoparticles. The cRGD-modified red blood cell membranes coated on the surface of the multidrug nanoparticles could enhance drug stability in circulation and tumor accumulation. This targeted combinational therapy significantly enhanced the antitumor efficiency in HER2-positive breast cancer in vitro and in vivo.
Collapse
Affiliation(s)
- Jie Xu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruichao Li
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Lijuan Zhu
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| |
Collapse
|
48
|
Jiao J, Qian Y, Lv Y, Wei W, Long Y, Guo X, Buerliesi A, Ye J, Han H, Li J, Zhu Y, Zhang W. Overcoming limitations and advancing the therapeutic potential of antibody-oligonucleotide conjugates (AOCs): Current status and future perspectives. Pharmacol Res 2024; 209:107469. [PMID: 39433169 DOI: 10.1016/j.phrs.2024.107469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
As cancer incidence rises due to an aging population, the importance of precision medicine continues to grow. Antibody-drug conjugates (ADCs) exemplify targeted therapies by delivering cytotoxic agents to specific antigens. Building on this concept, researchers have developed antibody-oligonucleotide conjugates (AOCs), which combine antibodies with oligonucleotides to regulate gene expression. This review highlights the mechanism of AOCs, emphasizing their unique ability to selectively target and modulate disease-causing proteins. It also explores the components of AOCs and their application in tumor therapy while addressing key challenges such as manufacturing complexities, endosomal escape, and immune response. The article underscores the significance of AOCs in precision oncology and discusses future directions, highlighting their potential in treating cancers driven by genetic mutations and abnormal protein expression.
Collapse
Affiliation(s)
- Jinlan Jiao
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Yun Qian
- Dermatologic Surgery Department, Institute of Dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing 210042, China
| | - Yinhua Lv
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Wenqian Wei
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Yongxuan Long
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Xiaoling Guo
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Anya Buerliesi
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Jiahui Ye
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Hao Han
- Department of Ultrasound, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jinbo Li
- State Key Laboratory of Analytical Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China.
| | - Yun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, China.
| | - Weijie Zhang
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China.
| |
Collapse
|
49
|
Lara P, Quiñonero F, Ortiz R, Prados J, Melguizo C. Nanoparticles Bounded to Interfering RNAs as a Therapy for Pancreatic Cancer: A Systematic Review. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2013. [PMID: 39510122 DOI: 10.1002/wnan.2013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Pancreatic cancer is one of the tumors with poor prognosis and low survival due to late diagnosis, high resistance, and very limited effective therapeutic options. Thus, new pharmacological treatments are necessary to improve the prognosis of patients. In this context, nanoparticles represent an efficient system for transporting and administering therapeutic molecules. Furthermore, siRNA can be used in cancer treatment to selectively inhibit the expression of any target gene. Therefore, nanoparticles associated with siRNA have been tested as a new therapeutic strategy to solve the pancreatic cancer treatment failure in the clinical setting. The current article presents a systematic revision of the literature of the last 10 years in which nanoparticles loading siRNA are used in pancreatic cancer. This research was carried out in three databases (PubMed, Scopus, and Web of Science) obtaining 164 articles from which 37 were selected. Our results show an overall view of the high effectiveness of this new therapy that combines nanoparticles with genetic therapy in pancreatic cancer suggesting that siRNA-based medicines will likely open up a new therapeutic era in the treatment of this type of tumors.
Collapse
Affiliation(s)
- Patricia Lara
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
| | - Francisco Quiñonero
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
| | - Raul Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
- Department of Anatomy and Embryology, University of Granada, Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
- Department of Anatomy and Embryology, University of Granada, Granada, Spain
| |
Collapse
|
50
|
Mrksich K, Padilla MS, Mitchell MJ. Breaking the final barrier: Evolution of cationic and ionizable lipid structure in lipid nanoparticles to escape the endosome. Adv Drug Deliv Rev 2024; 214:115446. [PMID: 39293650 PMCID: PMC11900896 DOI: 10.1016/j.addr.2024.115446] [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: 07/05/2024] [Revised: 08/18/2024] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
In the past decade, nucleic acid therapies have seen a boon in development and clinical translation largely due to advances in nanotechnology that have enabled their safe and targeted delivery. Nanoparticles can protect nucleic acids from degradation by serum enzymes and can facilitate entry into cells. Still, achieving endosomal escape to allow nucleic acids to enter the cytoplasm has remained a significant barrier, where less than 5% of nanoparticles within the endo-lysosomal pathway are able to transfer their cargo to the cytosol. Lipid-based drug delivery vehicles, particularly lipid nanoparticles (LNPs), have been optimized to achieve potent endosomal escape, and thus have been the vector of choice in the clinic as demonstrated by their utilization in the COVID-19 mRNA vaccines. The success of LNPs is in large part due to the rational design of lipids that can specifically overcome endosomal barriers. In this review, we chart the evolution of lipid structure from cationic lipids to ionizable lipids, focusing on structure-function relationships, with a focus on how they relate to endosomal escape. Additionally, we examine recent advancements in ionizable lipid structure as well as discuss the future of lipid design.
Collapse
Affiliation(s)
- Kaitlin Mrksich
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marshall S Padilla
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|