1
|
Gao Q, Wang W, Sun S, Yang Y, Mao K, Yang Y, Wu ZS. Bundling gold nanorods with RCA-produced DNA tape into an intelligently reconfigurable nanocluster bomb for multimodal precision cancer therapy. Mater Today Bio 2025; 32:101718. [PMID: 40236812 PMCID: PMC11999372 DOI: 10.1016/j.mtbio.2025.101718] [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: 01/30/2025] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/17/2025] Open
Abstract
Via proposing an innovative assembly technique, we bundle cell-targeting aptamer-modified gold nanorods (AuNRs) with RCA product (RCA-p) tape into a reconfigurable nanocluster (ARGN) bomb for multimodal precision cancer therapy. Because each ARGN has 10 individual AuNRs, the short time of laser irradiation can make the temperature increase to 75 °C much higher than the lethal temperature of tumor cells, enabling the efficient photothermal therapy (PTT). Moreover, both siRNA-Plk1 (2820 per ARGN) and chemotherapeutic agents (15860 per ARGN) can be loaded into two specifically-designed containers in the internal cavity. Because the glomeroplasmatic structure enhances the resistance to enzymatic degradation, ARGN bomb can protect siRNAs from the digestion and avoid Dox leakage during in vivo circulation. Moreover, the spontaneous structural reorganization allows aptamers in the interior cavity move outward to the exterior surface, which magically offers the compensation of degraded aptamers and impair persistent in vivo cell targeting ability. The external stimuli (laser irradiation) promotes the release of chemotherapeutic agents and initiates the PTT/chemotherapy outcome, while endogenous stimuli (intracellular biomarkers) causes almost 100 % release of siRNA-Plk1 species and induces RNA interference therapy, completely inhibiting tumor growth without detectable off-target toxicity.
Collapse
Affiliation(s)
- Qian Gao
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 305108, China
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Weijun Wang
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 305108, China
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- College of Chemistry and Food Science, Nanchang Normal University, Nanchang, 330032, China
| | - Shujuan Sun
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 305108, China
| | - Ya Yang
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 305108, China
| | - Kaili Mao
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuxi Yang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zai-Sheng Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 305108, China
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
2
|
Zhang Z, Fan YN, Jiang SQ, Ma YJ, Yu YR, Qing YX, Li QR, Liu YL, Shen S, Wang J. Recent Advances in mRNA Delivery Systems for Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e17571. [PMID: 40391789 DOI: 10.1002/advs.202417571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 04/01/2025] [Indexed: 05/22/2025]
Abstract
mRNA therapy is a promising approach in oncology, offering innovative applications such as tumor vaccines, protein replacement therapy, cell therapy, and gene therapy. However, challenges such as mRNA stability and delivery efficiency must be addressed. Advances in delivery system technologies are crucial for precise mRNA delivery, enhancing treatment safety and efficacy. The development of delivery systems requires accurate organ or cell targeting, intelligent release mechanisms, and optimized administration routes. This review outlines the applications of mRNA therapy in oncology, as well as the utilization of nonviral vectors, encompassing organic, inorganic, and biomimetic systems. It further elucidates the strategies for passive and active vector targeting and examines recent advances in the realm of stimuli-responsive delivery systems that are sensitive to pH and ultrasound. Additionally, the review addresses the development of noninvasive mRNA delivery systems designed for oral and pulmonary administration. The current challenges and emerging trends of mRNA therapy are discussed, and the potential strategies to mitigate these issues are emphasized.
Collapse
Affiliation(s)
- Zheng Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Ya-Nan Fan
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Si-Qi Jiang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Ya-Jing Ma
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Yao-Ru Yu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Yu-Xin Qing
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Qian-Ru Li
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Yi-Lin Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Song Shen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Jun Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
3
|
Jaiswal N, Mahata N, Chanda N. Nanogold-albumin conjugates: transformative approaches for next-generation cancer therapy and diagnostics. NANOSCALE 2025; 17:11191-11220. [PMID: 40237258 DOI: 10.1039/d4nr05279j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Nanogold-albumin conjugates have garnered significant attention as a highly adaptable theranostic platform, capable of delivering a wide range of therapeutics, from small-molecule drugs to larger biomolecules, while offering promising applications for monitoring and managing cancer. The remarkable theranostic capabilities of these conjugates stem from the combined strengths of gold and albumin, which provide low toxicity, a large surface area, customizable surface chemistry, and unique optical properties, all contributing to their potential in cancer therapy. This review delves into the design and development of two primary types of nanogold-albumin conjugate: supramolecular albumin-coated gold nanoparticles (GNP-BSA/HSA) and albumin-templated ultra-small gold nanoclusters (GNC-BSA/HSA). Each strategy offers distinct advantages, enabling the fine-tuning of conjugate properties to optimize therapeutic delivery and facilitate cancer-specific bio-sensing. The integration of gold and albumin further improves biocompatibility, extends circulation time, and enhances tumor targeting, making these conjugates an attractive option for cancer treatment. The review also focuses on the refinement of surface chemistry to achieve precise targeting of cancer cells, as well as the challenges and future prospects for advancing nanogold-albumin systems in clinical applications.
Collapse
Affiliation(s)
- Namita Jaiswal
- Human Centered Robotics and Cybernetics Group, CSIR-Central Mechanical Engineering Research Institute, Durgapur, India.
- Department of Biotechnology, National Institute of Technology (NIT), Durgapur, India
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology (NIT), Durgapur, India
| | - Nripen Chanda
- Human Centered Robotics and Cybernetics Group, CSIR-Central Mechanical Engineering Research Institute, Durgapur, India.
| |
Collapse
|
4
|
Ding L, Liu B, Peil A, Fan S, Chao J, Liu N. DNA‑Directed Assembly of Photonic Nanomaterials for Diagnostic and Therapeutic Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2500086. [PMID: 40103431 DOI: 10.1002/adma.202500086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/07/2025] [Indexed: 03/20/2025]
Abstract
DNA-directed assembly has emerged as a versatile and powerful approach for constructing complex structured materials. By leveraging the programmability of DNA nanotechnology, highly organized photonic systems can be developed to optimize light-matter interactions for improved diagnostics and therapeutic outcomes. These systems enable precise spatial arrangement of photonic components, minimizing material usage, and simplifying fabrication processes. DNA nanostructures, such as DNA origami, provide a robust platform for building multifunctional photonic devices with tailored optical properties. This review highlights recent progress in DNA-directed assembly of photonic nanomaterials, focusing on their applications in diagnostics and therapeutics. It provides an overview of the latest advancements in the field, discussing the principles of DNA-directed assembly, strategies for functionalizing photonic building blocks, innovations in assembly design, and the resulting optical effects that drive these developments. The review also explores how these photonic architectures contribute to diagnostic and therapeutic applications, emphasizing their potential to create efficient and effective photonic systems tailored to specific healthcare needs.
Collapse
Affiliation(s)
- Longjiang Ding
- 2nd Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
- Max Planck Institute for Solid State Research, Heisenbergstraße 1, 70569, Stuttgart, Germany
| | - Bing Liu
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Andreas Peil
- 2nd Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
- Max Planck Institute for Solid State Research, Heisenbergstraße 1, 70569, Stuttgart, Germany
| | - Sisi Fan
- 2nd Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
- Max Planck Institute for Solid State Research, Heisenbergstraße 1, 70569, Stuttgart, Germany
| | - Jie Chao
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Na Liu
- 2nd Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
- Max Planck Institute for Solid State Research, Heisenbergstraße 1, 70569, Stuttgart, Germany
| |
Collapse
|
5
|
Dykman L, Khlebtsov B, Khlebtsov N. Drug delivery using gold nanoparticles. Adv Drug Deliv Rev 2025; 216:115481. [PMID: 39617254 DOI: 10.1016/j.addr.2024.115481] [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: 07/12/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
Modern nanotechnologies provide various possibilities for efficiently delivering drugs to biological targets. This review focuses on using functionalized gold nanoparticles (GNPs) as a drug delivery platform. Owing to their exceptional size and surface characteristics, GNPs are a perfect drug delivery vehicle for targeted and selective distribution. Several in vitro and in vivo tests have shown how simple it is to tailor these particles to administer chemical medications straight to tumors. The GNP surface can also be coated with ligands to modify drug release or improve selectivity. Moreover, the pharmacological activity can be enhanced by using the photothermal characteristics of the particles.
Collapse
Affiliation(s)
- Lev Dykman
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - Boris Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - Nikolai Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia; Saratov State University, 83 Ulitsa Astrakhanskaya, Saratov 410012, Russia.
| |
Collapse
|
6
|
Yi X, Hussain I, Zhang P, Xiao C. Nuclear-Targeting Peptides for Cancer Therapy. Chembiochem 2024; 25:e202400596. [PMID: 39215136 DOI: 10.1002/cbic.202400596] [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: 07/15/2024] [Revised: 08/19/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Nucleus is the central regulator of cells that controls cell proliferation, metabolism, and cell cycle, and is considered the most important organelle in cells. The precision medicine that can achieve nuclear targeting has achieved good therapeutic effects in anti-tumor therapy. However, the presence of biological barriers such as cell membranes and nuclear membranes in cells limit the delivery of therapeutic agents to the nucleus. Therefore, developing effective nuclear-targeting drug delivery strategies is particularly important. Nuclear-targeting peptides are a class of functional peptides that can penetrate cell membranes and target the nucleus. They mainly recognize and bind to the nuclear transport molecules (such as Importin-α/β) and transport the therapeutic agents to the nucleus through nuclear pore complexes (NPC). This review summarizes the most recent developments of strategies for anti-tumor therapy utilizing nuclear-targeting peptides, which will ultimately contribute to the development of more effective nuclear-targeting strategies to achieve better anti-tumor outcomes.
Collapse
Affiliation(s)
- Xuan Yi
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China
| | - Irshad Hussain
- Department of Chemistry and Chemical Engineering, SBA School of Science & Engineering, Lahore University of Management Sciences (LUMS). DHA, Lahore, 54792, Pakistan
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China
| |
Collapse
|
7
|
He X, Tian Y, Dong J, Yuan Y, Zhang S, Jing H. RNA-Seq Reveals the Mechanism of Pyroptosis Induced by Oxygen-Enriched IR780 Nanobubbles-Mediated Sono-Photodynamic Therapy. Int J Nanomedicine 2024; 19:13029-13045. [PMID: 39654803 PMCID: PMC11625641 DOI: 10.2147/ijn.s487412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/23/2024] [Indexed: 12/12/2024] Open
Abstract
Background Sono-photodynamic therapy (SPDT), the combination of sonodynamic therapy (SDT) and photodynamic therapy (PDT), is a promising tumor treatment method. However, the hypoxic tumor microenvironment greatly compromises the efficacy of SPDT. Pyroptosis, a new type of programmed cell death, is mainly induced by some chemotherapeutic drugs in the current research, and rarely by SPDT. RNA sequencing (RNA-seq) is a high-throughput sequencing technique that comprehensively profiles the transcriptome, revealing the full spectrum of RNA molecules in a cell. Here, we constructed IR780@O2 nanobubbles (NBs) with photoacoustic dual response and hypoxia improvement properties to fight triple negative breast cancer (TNBC), and demonstrated that SPDT could kill TNBC cells through pyroptosis pathway. RNA-seq further revealed potential mechanisms and related differentially expressed genes. Methods Thin-film hydration and mechanical vibration method were utilized to synthesize IR780@O2 NBs. Subsequently, we characterized IR780@O2 NBs and examined the cytotoxicity as well as ROS production ability. A series of experiments were conducted to verify that SPDT killed TNBC cells through pyroptosis. Results IR780@O2 NBs were successfully prepared and had certain stability. Compared with SDT alone, SPDT increased therapeutic effect by 1.67 times by generating more ROS, and the introduction of NBs and O2 NBs (2.23 times and 2.93 times compared with SDT alone) could further promote this process. Other experiments proved that TNBC cells died by pyroptosis pathway. Moreover, the in-depth mechanism revealed that colony stimulating factor (CSF) and C-X-C motif chemokine ligand (CXCL) could be potential targets for the occurrence of pyroptosis in TNBC cells. Conclusion The IR780@O2 NBs prepared in this study increased the degree of TNBC cell pyroptosis through SPDT effect and alleviation of hypoxia, and cellular senescence might be a biological process closely related to pyroptosis in TNBC.
Collapse
Affiliation(s)
- Xiang He
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Jialin Dong
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yanchi Yuan
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Shijie Zhang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Hui Jing
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| |
Collapse
|
8
|
Peng X, Liu Y, Peng F, Wang T, Cheng Z, Chen Q, Li M, Xu L, Man Y, Zhang Z, Tan Y, Liu Z. Aptamer-controlled stimuli-responsive drug release. Int J Biol Macromol 2024; 279:135353. [PMID: 39245104 DOI: 10.1016/j.ijbiomac.2024.135353] [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: 06/12/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Aptamers have been widely researched and applied in nanomedicine due to their programmable, activatable, and switchable properties. However, there are few reviews on aptamer-controlled stimuli-responsive drug delivery. This article highlights the mechanisms and advantages of aptamers in the construction of stimuli-responsive drug delivery systems. We summarize the assembly/reconfiguration mechanisms of aptamers in controlled release systems. The assembly and drug release strategies of drug delivery systems are illustrated. Specifically, we focus on the binding mechanisms to the target and the factors that induce/inhibit the binding to the stimuli, such as strand, pH, light, and temperature. The applications of aptamer-based stimuli-responsive drug release are elaborated. The challenges are discussed, and the future directions are proposed.
Collapse
Affiliation(s)
- Xingxing Peng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Feicheng Peng
- Hunan Institute for Drug Control, Changsha 410001, Hunan Province, PR China
| | - Ting Wang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Zhongyu Cheng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yunqi Man
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Zhirou Zhang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China; Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan, PR China.
| |
Collapse
|
9
|
He X, Zhang S, Tian Y, Dong J, Yuan Y, Jing H. Nanobubble-mediated co-delivery of siTRIM37 and IR780 for gene and sonodynamic combination therapy against triple negative breast cancer. Biomed Mater 2024; 19:065015. [PMID: 39312939 DOI: 10.1088/1748-605x/ad7e6d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/23/2024] [Indexed: 09/25/2024]
Abstract
Gene therapy often fails due to enzyme degradation and low transfection efficiency, and single gene therapy usually cannot completely kill tumor cells. Several studies have reported that tripartite motif-containing protein 37 (TRIM37) plays a significant role in promoting the occurrence and development of triple negative breast cancer (TNBC). Herein, we constructed siTRIM37 and IR780 co-loaded nanobubbles (NBs) to achieve the combination of gene therapy and sonodynamic therapy (SDT) against TNBC. On the one hand, ultrasound irradiation causes siRNA@IR780 NBs rupture to produce ultrasound targeted NB destruction effect, which promotes the entry of IR780 and siTRIM37 into cells, increasing the local concentration of IR780 and gene transfection efficiency. On the other hand, under the stimulation of ultrasound, IR780 generates reactive oxygen species to kill TNBC cells. Mechanism studies reveal that TRIM37 is an anti-apoptotic gene in TNBC, and inhibiting TRIM37 expression can induce cell death through the apoptotic pathway. And the combination of siTRIM37 and SDT can aggravate the degree of apoptosis to increase cell death. Therefore, siRNA@IR780 NBs-mediated combination therapy may provide a new treatment approach for TNBC in the future.
Collapse
Affiliation(s)
- Xiang He
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, People's Republic of China
| | - Shentao Zhang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, People's Republic of China
| | - Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, People's Republic of China
| | - Jialin Dong
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, People's Republic of China
| | - Yanchi Yuan
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, People's Republic of China
| | - Hui Jing
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, People's Republic of China
| |
Collapse
|
10
|
Qiu L, Sun M, Chen L, Jiang J, Fu Z, Wang Y, Bi Y, Guo Q, Bai H, Chen S, Gao L, Chang G. Iron-Confined CRISPR/Cas9-Ribonucleoprotein Delivery System for Redox-Responsive Gene Editing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309431. [PMID: 38402425 DOI: 10.1002/smll.202309431] [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: 10/22/2023] [Revised: 02/08/2024] [Indexed: 02/26/2024]
Abstract
Clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein 9 (Cas9) is a promising gene editing tool to treat diseases at the genetic level. Nonetheless, the challenge of the safe and efficient delivery of CRISPR/Cas9 to host cells constrains its clinical applicability. In the current study, a facile, redox-responsive CRISPR/Cas9-Ribonucleoprotein (RNP) delivery system by combining iron-coordinated aggregation with liposomes (Fe-RNP@L) is reported. The Fe-RNP is formed by the coordination of Fe3+ with amino and carboxyl groups of Cas9, which modifies the lipophilicity and surface charge of RNP and alters cellular uptake from primary endocytosis to endocytosis and cholesterol-dependent membrane fusion. RNP can be rapidly and reversibly released from Fe-RNP in response to glutathione without loss of structural integrity and enzymatic activity. In addition, iron coordination also improves the stability of RNP and substantially mitigates cytotoxicity. This construct enabled highly efficient cytoplasmic/nuclear delivery (≈90%) and gene-editing efficiency (≈70%) even at low concentrations. The high payload content, high editing efficiency, good stability, low immunogenicity, and ease of production and storage, highlight its potential for diverse genome editing and clinical applications.
Collapse
Affiliation(s)
- Lingling Qiu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Minmin Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Lei Chen
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jing Jiang
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhendong Fu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Ying Wang
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Engineering Medicine, Beihang University, Beijing, China
| | - Yulin Bi
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Qixin Guo
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Hao Bai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Shihao Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guobin Chang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| |
Collapse
|
11
|
Tang F, Ding A, Xu Y, Ye Y, Li L, Xie R, Huang W. Gene and Photothermal Combination Therapy: Principle, Materials, and Amplified Anticancer Intervention. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307078. [PMID: 37775950 DOI: 10.1002/smll.202307078] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/19/2023] [Indexed: 10/01/2023]
Abstract
Gene therapy (GT) and photothermal therapy (PTT) have emerged as promising alternatives to chemotherapy and radiotherapy for cancer treatment, offering noninvasiveness and reduced side effects. However, their efficacy as standalone treatments is limited. GT exhibits slow response rates, while PTT is confined to local tumor ablation. The convergence of GT and PTT, known as GT-PTT, facilitated by photothermal gene nanocarriers, has attracted considerable attention across various disciplines. In this integrated approach, GT reciprocates PTT by sensitizing cellular response to heat, while PTT benefits GT by improving gene translocation, unpacking, and expression. Consequently, this integration presents a unique opportunity for cancer therapy with rapid response and improved effectiveness. Extensive efforts over the past few years have been dedicated to the development of GT-PTT, resulting in notable achievements and rapid progress from the laboratory to potential clinical applications. This comprehensive review outlines recent advances in GT-PTT, including synergistic mechanisms, material systems, imaging-guided therapy, and anticancer applications. It also explores the challenges and future prospects in this nascent field. By presenting innovative ideas and insights into the implementation of GT-PTT for enhanced cancer therapy, this review aims to inspire further progress in this promising area of research.
Collapse
Affiliation(s)
- Fang Tang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
- Future Display Institute in Xiamen, Xiamen, 361005, China
| | - Aixiang Ding
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
| | - Yao Xu
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
| | - Yingsong Ye
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
- Future Display Institute in Xiamen, Xiamen, 361005, China
- Frontiers Science Center for Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Rongjun Xie
- Fujian Key Laboratory of Materials Genome, College of Materials, Xiamen University, Xiamen, 361005, China
| | - Wei Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
- Future Display Institute in Xiamen, Xiamen, 361005, China
- Frontiers Science Center for Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| |
Collapse
|
12
|
Lin J, Wang X, Ni D, Chen Y, Chen C, Liu Y. Combinational Gene Therapy toward Cancer with Nanoplatform: Strategies and Principles. ACS MATERIALS AU 2023; 3:584-599. [PMID: 38089659 PMCID: PMC10636764 DOI: 10.1021/acsmaterialsau.3c00035] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 12/18/2024]
Abstract
Cancer remains a significant threat to human health. While numerous therapies have been developed to combat the disease, traditional treatments such as chemotherapy and radiotherapy are suboptimal and associated with significant side effects. Gene therapy is an emerging therapeutic approach that offers improved targeting and reduced side effects compared with traditional treatments. Using siRNA and other nucleic acid-based drugs in cancer treatment has generated significant interest among researchers. Nanocarriers, such as liposomes, can effectively deliver these agents to tumor sites. However, gene therapy alone is often insufficient to eradicate tumors, and there is a risk of recurrence. Therefore, combining gene therapy with other therapies using nanocarriers, such as phototherapy and magnetic hyperthermia therapy, can lead to synergistic therapeutic effects through different mechanisms. In this review, we summarize various ways in which gene therapy can be combined with other therapies and highlight the role of nanoplatforms in mediating these combined therapies, which would inspire novel design ideas toward combination therapies. Additionally, bottlenecks and barriers to gene therapy should be addressed in the near future to achieve better clinical efficacy.
Collapse
Affiliation(s)
- Jinhui Lin
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University
of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xinlian Wang
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University
of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Dongqi Ni
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University
of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Yandong Chen
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
| | - Chunying Chen
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University
of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Ying Liu
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
| |
Collapse
|
13
|
Chen S, Cao R, Xiang L, Li Z, Chen H, Zhang J, Feng X. Research progress in nucleus-targeted tumor therapy. Biomater Sci 2023; 11:6436-6456. [PMID: 37609783 DOI: 10.1039/d3bm01116j] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The nucleus is considered the most important organelle in the cell as it plays a central role in controlling cell reproduction, metabolism, and the cell cycle. The successful delivery of drugs into the nucleus can achieve excellent therapeutic effects, which reveals the potential of nucleus-targeted therapy in precision medicine. However, the transportation of therapeutics into the nucleus remains a significant challenge due to various biological barriers. Herein, we summarize the recent progress in the nucleus-targeted drug delivery system (NDDS). The structures of the nucleus and nuclear envelope are first described in order to understand the mechanisms by which drugs cross the nuclear envelope. Then, various drug delivery strategies based on the mechanisms and their applications are discussed. Finally, the challenges and solutions in the field of nucleus-targeted drug delivery are raised for developing a more efficient NDDS and promoting its clinical transformation.
Collapse
Affiliation(s)
- Shaofeng Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Rumeng Cao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Ling Xiang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Ziyi Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Hui Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Jiumeng Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Xuli Feng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| |
Collapse
|
14
|
Huang H, Liu R, Yang J, Dai J, Fan S, Pi J, Wei Y, Guo X. Gold Nanoparticles: Construction for Drug Delivery and Application in Cancer Immunotherapy. Pharmaceutics 2023; 15:1868. [PMID: 37514054 PMCID: PMC10383270 DOI: 10.3390/pharmaceutics15071868] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 05/28/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer immunotherapy is an innovative treatment strategy to enhance the ability of the immune system to recognize and eliminate cancer cells. However, dose limitations, low response rates, and adverse immune events pose significant challenges. To address these limitations, gold nanoparticles (AuNPs) have been explored as immunotherapeutic drug carriers owing to their stability, surface versatility, and excellent optical properties. This review provides an overview of the advanced synthesis routes for AuNPs and their utilization as drug carriers to improve precision therapies. The review also emphasises various aspects of AuNP-based immunotherapy, including drug loading, targeting strategies, and drug release mechanisms. The application of AuNPs combined with cancer immunotherapy and their therapeutic efficacy are briefly discussed. Overall, we aimed to provide a recent understanding of the advances, challenges, and prospects of AuNPs for anticancer applications.
Collapse
Affiliation(s)
- Huiqun Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Ronghui Liu
- School of Microelectronic, Southern University of Science and Technology, Shenzhen 518000, China
| | - Jie Yang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Jing Dai
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Shuhao Fan
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Jiang Pi
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Yubo Wei
- Yunnan Key Laboratory of Pharmacology for Natural Products, School of Pharmaceutical Sciences, Kunming Medical University, Kunming 650500, China
| | - Xinrong Guo
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
15
|
Mosley RJ, Rucci B, Byrne ME. Recent advancements in design of nucleic acid nanocarriers for controlled drug delivery. J Mater Chem B 2023; 11:2078-2094. [PMID: 36806872 DOI: 10.1039/d2tb02325c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Research of nanoscale nucleic acid carriers has garnered attention in recent years due to their distinctive and controllable properties. However, current knowledge is limited in how we can efficiently utilize these systems for clinical applications. Several researchers have pioneered new and innovative nanocarrier drug delivery systems, but understanding physiochemical properties and behavior in vivo is vital to implementing them as clinical drug delivery platforms. In this review, we outline the most significant innovations in the synthesis, physical properties, and utilization of nucleic acid nanocarriers in the past 5 years, addressing the crucial properties which improve nanocarrier characteristics, delivery, and drug release. The challenges of controlling the transport of nucleic acid nanocarriers and therapeutic release for biological applications are outlined. Barriers which inhibit effective transport into tissue are discussed with emphasis on the modifications needed to overcome such obstacles. The novel strategies discussed in this work summarize the pivotal features of modern nucleic nanocarriers and postulate where future developments could revolutionize the translation of these tools into a clinical setting.
Collapse
Affiliation(s)
- Robert J Mosley
- Biomimetic and Biohybrid Materials, Biomedical Devices, and Drug Delivery Laboratories, Department of Biomedical Engineering, 201 Mullica Hill Rd, Rowan University, Glassboro, NJ, 08028, USA.
| | - Brendan Rucci
- Biomimetic and Biohybrid Materials, Biomedical Devices, and Drug Delivery Laboratories, Department of Biomedical Engineering, 201 Mullica Hill Rd, Rowan University, Glassboro, NJ, 08028, USA.
| | - Mark E Byrne
- Biomimetic and Biohybrid Materials, Biomedical Devices, and Drug Delivery Laboratories, Department of Biomedical Engineering, 201 Mullica Hill Rd, Rowan University, Glassboro, NJ, 08028, USA. .,Department of Chemical Engineering, Rowan University, Glassboro, NJ, 08028, USA
| |
Collapse
|
16
|
Sobhanan J, Anas A, Biju V. Nanomaterials for Fluorescence and Multimodal Bioimaging. CHEM REC 2023; 23:e202200253. [PMID: 36789795 DOI: 10.1002/tcr.202200253] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/27/2023] [Indexed: 02/16/2023]
Abstract
Bioconjugated nanomaterials replace molecular probes in bioanalysis and bioimaging in vitro and in vivo. Nanoparticles of silica, metals, semiconductors, polymers, and supramolecular systems, conjugated with contrast agents and drugs for image-guided (MRI, fluorescence, PET, Raman, SPECT, photodynamic, photothermal, and photoacoustic) therapy infiltrate into preclinical and clinical settings. Small bioactive molecules like peptides, proteins, or DNA conjugated to the surfaces of drugs or probes help us to interface them with cells and tissues. Nevertheless, the toxicity and pharmacokinetics of nanodrugs, nanoprobes, and their components become the clinical barriers, underscoring the significance of developing biocompatible next-generation drugs and contrast agents. This account provides state-of-the-art advancements in the preparation and biological applications of bioconjugated nanomaterials and their molecular, cell, and in vivo applications. It focuses on the preparation, bioimaging, and bioanalytical applications of monomodal and multimodal nanoprobes composed of quantum dots, quantum clusters, iron oxide nanoparticles, and a few rare earth metal ion complexes.
Collapse
Affiliation(s)
- Jeladhara Sobhanan
- Graduate School of Environmental Science, Hokkaido University, N10 W5, Sapporo, Hokkaido, 060-0810, Japan.,Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Abdulaziz Anas
- CSIR-National Institute of Oceanography, Regional Centre Kochi, Kerala, 682 018, India
| | - Vasudevanpillai Biju
- Graduate School of Environmental Science, Hokkaido University, N10 W5, Sapporo, Hokkaido, 060-0810, Japan.,Research Institute for Electronic Science, Hokkaido University, Sapporo, 001-0020, Japan
| |
Collapse
|
17
|
Ouyang R, Zhang Q, Cao P, Yang Y, Zhao Y, Liu B, Miao Y, Zhou S. Efficient improvement in chemo/photothermal synergistic therapy against lung cancer using Bi@Au nano-acanthospheres. Colloids Surf B Biointerfaces 2023; 222:113116. [PMID: 36603409 DOI: 10.1016/j.colsurfb.2022.113116] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022]
Abstract
Novel highly hydrophilic and biocompatible bismuth nanospheres with gold nanoparticles growing outside (Bi@Au nano-acanthospheres, Bi@Au NASs) were synthesized through a simple procedure, which demonstrated to be a promising photothermal agent owing to the ultrahigh photothermal conversion efficiency (η = 46.6 %). The as-prepared Bi@Au NASs showed excellent blood compatibility and fairly low cytotoxicity to human lung cancer A549 cells, as well as efficient photothermal ablation (PTA) therapy induced by a near-infrared laser. Under the 808 nm laser radiation, the tumour temperature could be elevated by ∼25 °C high enough to kill the cancer cells. Moreover, the anticancer drug doxorubicin hydrochloride (DOX) was successfully loaded in Bi@Au NASs with a loading content as high as 16.78 % and released under a pH sensitive release profile, a characteristic beneficial for intravenous delivery of DOX into cancer cells for chemotherapy. The presence of the Bi element enabled Bi@Au NASs to act as a favourable computed tomography (CT) contrast medium for CT imaging-guided tumour treatment. Compared with cancer treatment through either photothermal therapy or chemotherapy, the chemo-photothermal synergistic therapy using Bi@Au NASs as both a photothermal agent and a drug carrier has efficiently enhanced the in vitro and in vivo therapeutic effects in cancer treatment.
Collapse
Affiliation(s)
- Ruizhuo Ouyang
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Qiupeng Zhang
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Penghui Cao
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yang Yang
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yuefeng Zhao
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Baolin Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yuqing Miao
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Shuang Zhou
- Cancer Institute, Tongji University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
18
|
Gao Y, Wang K, Zhang J, Duan X, Sun Q, Men K. Multifunctional nanoparticle for cancer therapy. MedComm (Beijing) 2023; 4:e187. [PMID: 36654533 PMCID: PMC9834710 DOI: 10.1002/mco2.187] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 01/14/2023] Open
Abstract
Cancer is a complex disease associated with a combination of abnormal physiological process and exhibiting dysfunctions in multiple systems. To provide effective treatment and diagnosis for cancer, current treatment strategies simultaneously focus on various tumor targets. Based on the rapid development of nanotechnology, nanocarriers have been shown to exhibit excellent potential for cancer therapy. Compared with nanoparticles with single functions, multifunctional nanoparticles are believed to be more aggressive and potent in the context of tumor targeting. However, the development of multifunctional nanoparticles is not simply an upgraded version of the original function, but involves a sophisticated system with a proper backbone, optimized modification sites, simple preparation method, and efficient function integration. Despite this, many well-designed multifunctional nanoparticles with promising therapeutic potential have emerged recently. Here, to give a detailed understanding and analyzation of the currently developed multifunctional nanoparticles, their platform structures with organic or inorganic backbones were systemically generalized. We emphasized on the functionalization and modification strategies, which provide additional functions to the nanoparticle. We also discussed the application combination strategies that were involved in the development of nanoformulations with functional crosstalk. This review thus provides an overview of the construction strategies and application advances of multifunctional nanoparticles.
Collapse
Affiliation(s)
- Yan Gao
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Kaiyu Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Xingmei Duan
- Department of PharmacyPersonalized Drug Therapy Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuan ProvinceChina
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| |
Collapse
|
19
|
He X, Zhang S, Tian Y, Cheng W, Jing H. Research Progress of Nanomedicine-Based Mild Photothermal Therapy in Tumor. Int J Nanomedicine 2023; 18:1433-1468. [PMID: 36992822 PMCID: PMC10042261 DOI: 10.2147/ijn.s405020] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
With the booming development of nanomedicine, mild photothermal therapy (mPTT, 42-45°C) has exhibited promising potential in tumor therapy. Compared with traditional PTT (>50°C), mPTT has less side effects and better biological effects conducive to tumor treatment, such as loosening the dense structure in tumor tissues, enhancing blood perfusion, and improving the immunosuppressive microenvironment. However, such a relatively low temperature cannot allow mPTT to completely eradicate tumors, and therefore, substantial efforts have been conducted to optimize the application of mPTT in tumor therapy. This review extensively summarizes the latest advances of mPTT, including two sections: (1) taking mPTT as a leading role to maximize its effect by blocking the cell defense mechanisms, and (2) regarding mPTT as a supporting role to assist other therapies to achieve synergistic antitumor curative effect. Meanwhile, the special characteristics and imaging capabilities of nanoplatforms applied in various therapies are discussed. At last, this paper puts forward the bottlenecks and challenges in the current research path of mPTT, and possible solutions and research directions in future are proposed correspondingly.
Collapse
Affiliation(s)
- Xiang He
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Shentao Zhang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Hui Jing
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
- Correspondence: Hui Jing; Wen Cheng, Department of Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, People’s Republic of China, Tel +86 13304504935; +86 13313677182, Email ;
| |
Collapse
|
20
|
Fang J, Feng Y, Zhang Y, Wang A, Li J, Cui C, Guo Y, Zhu J, Lv Z, Zhao Z, Xu C, Shi H. Alkaline Phosphatase-Controllable and Red Light-Activated RNA Modification Approach for Precise Tumor Suppression. J Am Chem Soc 2022; 144:23061-23072. [DOI: 10.1021/jacs.2c10409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jing Fang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yali Feng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yuqi Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Anna Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jiachen Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Chaoxiang Cui
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Yirui Guo
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jinfeng Zhu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Zhengzhong Lv
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Zhongsheng Zhao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Haibin Shi
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
21
|
Jiang Y, Fan M, Yang Z, Liu X, Xu Z, Liu S, Feng G, Tang S, Li Z, Zhang Y, Chen S, Yang C, Law WC, Dong B, Xu G, Yong KT. Recent advances in nanotechnology approaches for non-viral gene therapy. Biomater Sci 2022; 10:6862-6892. [PMID: 36222758 DOI: 10.1039/d2bm01001a] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gene therapy has shown great potential in the treatment of many diseases by downregulating the expression of certain genes. The development of gene vectors as a vehicle for gene therapy has greatly facilitated the widespread clinical application of nucleic acid materials (DNA, mRNA, siRNA, and miRNA). Currently, both viral and non-viral vectors are used as delivery systems of nucleic acid materials for gene therapy. However, viral vector-based gene therapy has several limitations, including immunogenicity and carcinogenesis caused by the exogenous viral vectors. To address these issues, non-viral nanocarrier-based gene therapy has been explored for superior performance with enhanced gene stability, high treatment efficiency, improved tumor-targeting, and better biocompatibility. In this review, we discuss various non-viral vector-mediated gene therapy approaches using multifunctional biodegradable or non-biodegradable nanocarriers, including polymer-based nanoparticles, lipid-based nanoparticles, carbon nanotubes, gold nanoparticles (AuNPs), quantum dots (QDs), silica nanoparticles, metal-based nanoparticles and two-dimensional nanocarriers. Various strategies to construct non-viral nanocarriers based on their delivery efficiency of targeted genes will be introduced. Subsequently, we discuss the cellular uptake pathways of non-viral nanocarriers. In addition, multifunctional gene therapy based on non-viral nanocarriers is summarized, in which the gene therapy can be combined with other treatments, such as photothermal therapy (PTT), photodynamic therapy (PDT), immunotherapy and chemotherapy. We also provide a comprehensive discussion of the biological toxicity and safety of non-viral vector-based gene therapy. Finally, the present limitations and challenges of non-viral nanocarriers for gene therapy in future clinical research are discussed, to promote wider clinical applications of non-viral vector-based gene therapy.
Collapse
Affiliation(s)
- Yihang Jiang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Miaozhuang Fan
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Zhenxu Yang
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia. .,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia.,The Biophotonics and Mechanobioengineering Laboratory, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Xiaochen Liu
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia. .,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia.,The Biophotonics and Mechanobioengineering Laboratory, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Zhourui Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Shikang Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Gang Feng
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Shuo Tang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Zhengzheng Li
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Yibin Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Shilin Chen
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Wing-Cheung Law
- Department of Industrial and Systems Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon 999077, Hong Kong, China
| | - Biqin Dong
- Guangdong Provincial Key Laboratory of Durability for Marine Civil Engineering, College of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia. .,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia.,The Biophotonics and Mechanobioengineering Laboratory, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
22
|
Xu R, Li Y, Zhu C, Liu D, Yang YR. Cellular Ingestible DNA Nanostructures for Biomedical Applications. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Rui Xu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 China
| | - Yujie Li
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 China
| | - Chenyou Zhu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 China
| | - Dongsheng Liu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 China
| | - Yuhe R. Yang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing 100190 China
| |
Collapse
|
23
|
Intelligent nanotherapeutic strategies for the delivery of CRISPR system. Acta Pharm Sin B 2022. [DOI: 10.1016/j.apsb.2022.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
24
|
Fang T, Cao X, Ibnat M, Chen G. Stimuli-responsive nanoformulations for CRISPR-Cas9 genome editing. J Nanobiotechnology 2022; 20:354. [PMID: 35918694 PMCID: PMC9344766 DOI: 10.1186/s12951-022-01570-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/22/2022] [Indexed: 12/07/2022] Open
Abstract
The CRISPR-Cas9 technology has changed the landscape of genome editing and has demonstrated extraordinary potential for treating otherwise incurable diseases. Engineering strategies to enable efficient intracellular delivery of CRISPR-Cas9 components has been a central theme for broadening the impact of the CRISPR-Cas9 technology. Various non-viral delivery systems for CRISPR-Cas9 have been investigated given their favorable safety profiles over viral systems. Many recent efforts have been focused on the development of stimuli-responsive non-viral CRISPR-Cas9 delivery systems, with the goal of achieving efficient and precise genome editing. Stimuli-responsive nanoplatforms are capable of sensing and responding to particular triggers, such as innate biological cues and external stimuli, for controlled CRISPR-Cas9 genome editing. In this Review, we overview the recent advances in stimuli-responsive nanoformulations for CRISPR-Cas9 delivery, highlight the rationale of stimuli and formulation designs, and summarize their biomedical applications.
Collapse
Affiliation(s)
- Tianxu Fang
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Xiaona Cao
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada.,School of Nursing, Tianjin Medical University, Tianjin, China
| | - Mysha Ibnat
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada. .,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada.
| |
Collapse
|
25
|
Jaiswal N, Halder S, Mahata N, Chanda N. Bi-Functional Gold Nanorod-Protein Conjugates with Biomimetic BSA@Folic Acid Corona for Improved Tumor Targeting and Intracellular Delivery of Therapeutic Proteins in Colon Cancer 3D Spheroids. ACS APPLIED BIO MATERIALS 2022; 5:1476-1488. [PMID: 35285613 DOI: 10.1021/acsabm.1c01216] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Gold nanorods (AuNRs) remain well-developed inorganic nanocarriers of small molecules for a plethora of biomedical and therapeutic applications. However, the delivery of therapeutic proteins using AuNRs with high protein loading capacity (LC), serum stability, excellent target specificity, and minimal off-target protein release is not known. Herein, we report two bi-functional AuNR-protein nanoconjugates, AuNR@EGFP-BSAFA and AuNR@RNaseA-BSAFA, supramolecularly coated with folic acid-modified BSA (BSAFA) acting as biomimetic protein corona to demonstrate targeted cytosolic delivery of enhanced green fluorescent protein (EGFP) and therapeutic ribonuclease A enzyme (RNase A) in their functional forms. AuNR@EGFP-BSAFA and AuNR@RNaseA-BSAFA exhibit high LCs of ∼42 and ∼54%, respectively, increased colloidal stability, and rapid protein release in the presence of biological thiols. As a nanocarrier, AuNR@EGFP-BSAFA and AuNR@RNaseA-BSAFA show resistance to corona formation in high-serum media even after 24 h, guaranteeing a greater circulation lifetime. Folate receptor-targeting BSAFA on the AuNR surface facilitates the receptor-mediated internalization, followed by the release of EGFP and RNase A in HT29 cells. The green fluorescence dispersed throughout the cell's cytoplasm indicates successful cytosolic delivery of EGFP by AuNR@EGFP-BSAFA. AuNR@RNaseA-BSAFA-mediated therapeutic RNase A delivery in multicellular 3D spheroids of HT29 cells exhibits a radical reduction in the cellular RNA fluorescence intensity to 38%, signifying RNA degradation and subsequent cell death. The versatile nanoformulation strategy in terms of the anisotropic particle morphology, protein type, and ability for targeted delivery in the functional form makes the present AuNR-protein nanoconjugates a promising platform for potential application in cancer management.
Collapse
Affiliation(s)
- Namita Jaiswal
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713209, India.,Material Processing and Microsystem Laboratory, CSIR─Central Mechanical Engineering Research Institute, Durgapur 713209, India
| | - Sudeshna Halder
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713209, India
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713209, India
| | - Nripen Chanda
- Material Processing and Microsystem Laboratory, CSIR─Central Mechanical Engineering Research Institute, Durgapur 713209, India
| |
Collapse
|
26
|
Tang N, Ning Q, Wang Z, Tao Y, Zhao X, Tang S. Tumor microenvironment based stimuli-responsive CRISPR/Cas delivery systems: A viable platform for interventional approaches. Colloids Surf B Biointerfaces 2021; 210:112257. [PMID: 34894597 DOI: 10.1016/j.colsurfb.2021.112257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023]
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas) systems have emerged as robust tools in cancer gene therapy due to their simplicity and versatility. Nevertheless, the genome editing efficiency in tumor sites and the clinical applications of CRISPR/Cas have been compromised by non-specific delivery and genotoxicity. Recently, intelligent delivery systems incorporating sensitive materials in response to endogenous stimuli of the tumor microenvironment (TME) have represented viable platforms for tumor-specific genome editing and reduced side effects of CRISPR/Cas. Spurred by this promising direction, this review first introduces the CRISPR/Cas systems widely employed in cancer therapeutic explorations. Various types of CRISPR/Cas delivery systems sensitive to the stimuli in TME and typical dual-/multiple-responsive CRISPR/Cas carriers are further discussed, emphasizing the correlations between sensitive components and spatiotemporal delivery mechanisms. The genome editing efficiencies of CRISPR/Cas-loaded stimuli-responsive carriers are also summarized both in vitro and in vivo. Collectively, stimuli-responsive CRISPR/Cas delivery systems hold great promise for potent cancer gene therapy.
Collapse
Affiliation(s)
- Nanyang Tang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, and Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China.
| | - Qian Ning
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| | - Zewei Wang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, and Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China.
| | - Yifang Tao
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, and Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China.
| | - Xuhong Zhao
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China.
| | - Shengsong Tang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, and Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|