1
|
Gong Q, Song X, Tong Y, Huo L, Zhao X, Han Y, Shen W, Ru J, Shen X, Liang C. Recent advances of anti-tumor nano-strategies via overturning pH gradient: alkalization and acidification. J Nanobiotechnology 2025; 23:42. [PMID: 39849540 PMCID: PMC11761731 DOI: 10.1186/s12951-025-03134-2] [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: 10/17/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025] Open
Abstract
The acidic tumor microenvironment, a hallmark of many solid tumors, is primarily induced by the high glycolytic rate of tumor cells. To avoid acidosis, tumor cells ingeniously maintain an acidic extracellular pH while keeping a relatively alkaline intracellular pH. Overturning the unique pH gradient of tumor cells has exhibited to be a viable approach for cancer therapy. In this review, the formation and regulatory mechanisms of the acidic microenvironment in solid tumors will be firstly outlined. Subsequently, we will comprehensively summarize the latest advancements in anti-tumor therapy via using nanomedicines to manipulate the tumor pH gradient, including acidifying intracellular environment and alkalizing extracellular environment. Following this, we will discuss the future potential of strategies employing nanomedicines to reverse tumor pH gradient. This review aims to foster research on therapeutic approaches targeting the pH regulation of solid tumors and holds an optimistic outlook for the future development of this field.
Collapse
Affiliation(s)
- Qiufang Gong
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xuejiao Song
- School of Physical and Mathematical Sciences, Nanjing Tech University (Nanjing Tech), Nanjing, 211816, China.
| | - Yao Tong
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lixuan Huo
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xuefen Zhao
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yingying Han
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wei Shen
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jiaxi Ru
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xian Shen
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Chao Liang
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
2
|
Li M, Zhang H, Xiong P, He Y, Zhou W, Wu C, Liao X, Zhang W, Yang H, Liu Y. DNA origami-based composite nanosandwich for iteratively potentiated chemo-immunotherapy. J Control Release 2025; 379:452-465. [PMID: 39809421 DOI: 10.1016/j.jconrel.2025.01.031] [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: 08/25/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Developing effective nanoplatforms for chemo-immunotherapy to achieve enhanced tumor suppression and systemic antitumor immunity has recently received extensive attention. Herein, we formulated a multifunctional DNA sandwich nanodevice, DSWAC/siPD-L1, based on triangular DNA origami, to implement enhanced cancer chemo-immunotherapy. Taking advantage of the tumor-targeting ability of the AS1411 aptamer, DSWAC/siPD-L1 efficiently delivered doxorubicin (DOX), CpG, and siPD-L1 into tumor cells. Moreover, the sandwich cavity spatially protects siPD-L1 from degradation, and the featured design of the DNA/RNA duplex linkers ensures effective intracellular release of siPD-L1. The pH-responsive release of cytotoxic DOX induces apoptosis and initial mild immunogenic cell death of tumor cells, presenting antigens to enhance the maturation of dendritic cells (DCs) with the assistance of the immune adjuvant CpG, thereby activating cytotoxic T lymphocytes to amplify antitumor immunity. Simultaneously, siPD-L1 downregulated the endogenous expression of PD-L1 to inhibit adaptive tumor immune escape. DSWAC/siPD-L1 initiated the iterative revolution of the cancer-immunity cycle, leading to the inhibition of primary and metastatic tumors, as demonstrated by DC maturation and T-cell infiltration in established subcutaneous primary tumor model and metastatic lung tumor model. Furthermore, the superior antitumor effect of DSWAC/siPD-L1 resulted in approximately 91 % inhibition of primary tumor growth and 93 % prevention of lung metastasis. Collectively, this study describes a siPD-L1-based sandwich DNA nanodevice functionalized with AS1411/CpG for enhanced cancer chemo-immunotherapy, inspiring the creation of more innovative drug nanocarriers and the exploitation of novel cancer therapies.
Collapse
Affiliation(s)
- Mengyue Li
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Hanxi Zhang
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Peizheng Xiong
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, PR China
| | - Yuhan He
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Wanyi Zhou
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Chunhui Wu
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Xiaoling Liao
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, PR China.
| | - Wei Zhang
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China.
| | - Hong Yang
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China.
| | - Yiyao Liu
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, PR China; Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, PR China; Department of Urology, Deyang People's Hospital, Deyang 618099, Sichuan, PR China.
| |
Collapse
|
3
|
Chi Y, Song C, Jia Q, Zhang R, Sun F, Li Z, Jia Y, An X, Wang Z, Li J. A metal coordination polymer nanoparticle synergistically re-establishes acidosis and enhances chemodynamic therapy for Glioblastoma. Acta Biomater 2025; 192:290-301. [PMID: 39608659 DOI: 10.1016/j.actbio.2024.11.042] [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: 08/17/2024] [Revised: 11/12/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Chemodynamic therapy (CDT) has become increasingly important as a tumor treatment strategy, which relies on intracellular acid and hydrogen peroxide to kill tumor cells by generating hydroxyl radicals (·OH) through Fenton/Fenton-like reactions. However, the weakly alkaline intracellular environment considerably caused by the efflux of lactate and H+ from glioblastoma cells is not conducive to CDT performance. Intracellular acidification induced by inhibiting the transmembrane monocarboxylate transporter 4 (MCT4) can enhance the therapeutic efficacy of CDT. Existing approaches suffer from insufficient MCT4 inhibition, involve complex drug synthesis, and have many unsatisfactory side effects. METHODS In this study, we constructed an anti-tumor nanoparticle formed by self-assembly driven by the coordination interaction of Fe3+ and α-cyano-4-hydroxycinnamate (CHC) to avoid safety issues posed by excessive modification. Fe-CHC nanoparticles were designed to decrease intracellular pH through inhibition of MCT4, which transports lactate/H+ to the extracellular space. The resulting intracellular accumulation of lactate and H+ led to fatal acidosis and promoted ·OH generated by Fenton/Fenton-like reactions with the presence of the Fe3+, thus enhancing CDT-induced tumor cell death. RESULTS In vitro and in vivo results revealed that Fe-CHC exerted a significant synergistic anti-tumor effect by re-establishing acidosis and enhancing CDT in glioblastoma. Furthermore, the decreased H+outside the cells caused by the inhibition of lactate/H+ efflux hindered extracellular matrix degradation, thereby inhibiting tumor metastasis. CONCLUSION Fe-CHC is an effective anti-cancer agent against glioblastoma. This study provides valuable insights for developing acid-modulating anti-tumor nanoparticles, as well as enriching and optimizing the application of CDT in tumor therapy. STATEMENT OF SIGNIFICANCE Our study pioneers the Fe-CHC nanoparticle, a metal-coordination polymer that targets MCT4 in glioblastoma cells to restore intracellular acidity and synergize with Fe3+ to boost chemodynamic therapy (CDT). Unlike other studies, Fe3+ and CHC work together to maximize the therapeutic potential and safety of Fe-CHC with minimal complexity. This innovative approach not only increased the production of reactive oxygen species within tumor cells, but also hindered tumor metastasis. Our work has important scientific implications for tumor microenvironment regulation and the application of CDT, and will provide a promising pathway for the treatment of aggressive cancers and attract a wide audience through its scientific implications.
Collapse
Affiliation(s)
- Yajing Chi
- School of Medicine, Nankai University, Tianjin, 300071, China; Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Chaoqi Song
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Qian Jia
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China; Guangzhou Institute of Technology, Xidian University, Guangzhou, GuangDong, 510000, China.
| | - Ruili Zhang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Fang Sun
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Zheng Li
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Yuanyuan Jia
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Xian An
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing, 100071, China
| | - Zhongliang Wang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China.
| | - Jianxiong Li
- School of Medicine, Nankai University, Tianjin, 300071, China; Department of Radiotherapy, Chinese PLA General Hospital, Beijing, 100071, China.
| |
Collapse
|
4
|
Xue F, Zhao H, Liu H, Lou J, Li K, Wang Z, An L, Tian Q. Autophagic cell death induced by pH modulation for enhanced iron-based chemodynamic therapy. J Colloid Interface Sci 2025; 678:13-23. [PMID: 39276684 DOI: 10.1016/j.jcis.2024.09.093] [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: 07/24/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
Iron-based chemodynamic therapy (CDT) exhibits commendable biocompatibility and selectivity, but its efficacy is constrained by the intracellular pH of tumors. To overcome this obstacle, we constructed a silica delivery platform loaded with autophagy-inducing reagents (rapamycin, RAPA) and iron-based Fenton reagents (Fe3O4). This platform was utilized to explore a novel strategy that leverages autophagy to decrease tumor acidity, consequently boosting the effectiveness of CDT. Both in vitro and in vivo experiments revealed that RAPA prompted the generation of acidic organelles (e.g., autophagic vacuoles and autophagosomes), effectively changing the intracellular pH in the tumor microenvironment. Furthermore, RAPA-induced tumor acidification significantly amplified the efficacy of Fe3O4-based Fenton reactions, consequently increasing the effectiveness of Fe3O4-based CDT. This innovative approach, which leverages the interplay between autophagy induction and iron-based CDT, shows promise in overcoming the limitations posed by tumor pH, thus offering a more efficient approach to tumor treatments.
Collapse
Affiliation(s)
- Fengfeng Xue
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Huifeng Zhao
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Hui Liu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jingjing Lou
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China.
| | - Kailin Li
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Zikang Wang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Lu An
- The Education Ministry Key Lab of Resource Chemistry, Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China.
| | - Qiwei Tian
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| |
Collapse
|
5
|
Pan Y, Zhao H, Huang W, Liu S, Qi Y, Huang Y. Metal-Protein Hybrid Materials: Unlocking New Frontiers in Biomedical Applications. Adv Healthc Mater 2025:e2404405. [PMID: 39778029 DOI: 10.1002/adhm.202404405] [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: 11/07/2024] [Revised: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Metal-protein hybrid materials represent a novel class of functional materials that exhibit exceptional physicochemical properties and tunable structures, rendering them remarkable applications in diverse fields, including materials engineering, biocatalysis, biosensing, and biomedicine. The design and development of multifunctional and biocompatible metal-protein hybrid materials have been the subject of extensive research and a key aspiration for practical applications in clinical settings. This review provides a comprehensive analysis of the design strategies, intrinsic properties, and biomedical applications of these hybrid materials, with a specific emphasis on their potential in cancer therapy, drug and vaccine delivery, antibacterial treatments, and tissue regeneration. Through rational design, stable metal-protein hybrid materials can be synthesized using straightforward methods, enabling them with therapeutic, delivery, immunomodulatory, and other desired functionalities. Finally, the review outlines the existing limitations and challenges associated with metal-protein hybrid materials and evaluates their potential for clinical translation, providing insights into their practical implementation within biomedical applications.
Collapse
Affiliation(s)
- Yong Pan
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Han Zhao
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Wenyong Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Siyang Liu
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yanxin Qi
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| |
Collapse
|
6
|
Wang L, Zhang X, He L, Wei Y, Zhang Y, Wu A, Li J. Iron-Based Nanomaterials for Modulating Tumor Microenvironment. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70001. [PMID: 39788569 DOI: 10.1002/wnan.70001] [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: 07/06/2024] [Revised: 11/29/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025]
Abstract
Iron-based nanomaterials (IBNMs) have been widely applied in biomedicine applications including magnetic resonance imaging, targeted drug delivery, tumor therapy, and so forth, due to their unique magnetism, excellent biocompatibility, and diverse modalities. Further research on its enormous biomedical potential is still ongoing, and its new features are constantly being tapped and demonstrated. Among them, various types of IBNMs have demonstrated significant cancer therapy capabilities by regulating the tumor microenvironment (TME). In this review, a variety of IBNMs including iron oxide-based nanomaterials (IONMs), iron-based complex conjugates (ICCs), and iron-based single iron atom nanomaterials (ISANMs) will be introduced, and their advantages in regulating TME would also be emphasized. Besides, the recent progress of IBNMs for cancer diagnosis and treatment through the strategy of modulating TME will be summarized, including overcoming hypoxia, modulating acidity, decreasing redox species, and immunoregulation. Finally, the challenges and opportunities in this field are briefly discussed. This review is expected to contribute to the future design and development of next-generation TME-modulate IBNMs for cancer treatment.
Collapse
Affiliation(s)
- Le Wang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Xiaoting Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Lulu He
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Yuanyuan Wei
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Yujie Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Aiguo Wu
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Juan Li
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| |
Collapse
|
7
|
Gao Y, Li A, Li Y, Guo H, He L, Li K, Shcharbin D, Shi X, Shen M. Dendrimer/Copper(II) Complex-Mediated siRNA Delivery Disrupts Lactate Metabolism to Reprogram the Local Immune Microenvironment against Tumor Growth and Metastasis. Biomacromolecules 2024; 25:7995-8005. [PMID: 39570391 DOI: 10.1021/acs.biomac.4c01249] [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: 11/22/2024]
Abstract
Solid tumors reprogram metabolic pathways to meet their biosynthesis demands, resulting in elevated levels of metabolites in the tumor microenvironment (TME), including lactate. Excessive accumulation and active transportation of lactate within the TME drives tumor progression, metastasis, and immunosuppression. Interruption of TME lactate metabolism is expected to restore antitumor responses and sensitize tumor immunotherapy. Herein, we developed phenylboronic acid- and pyridine-modified poly(amidoamine) dendrimer/copper(II) (Cu(II)) complexes, namely, D-Cu complexes, to deliver monocarboxylate transporter 4 siRNA (siMCT4) and disrupt the tumor lactate shuttle. The D-Cu complexes are shown to have a Cu(II)-mediated chemodynamic effect and T1-weighted magnetic resonance imaging potential (r1 relaxivity = 1.19 mM-1 s-1), enabling effective siMCT4 delivery to inhibit lactate efflux within cancer cells. In combination with a CD11b immune agonist, the treatment of D-Cu/siMCT4 polyplexes in a mouse breast tumor model alleviates local TME immunosuppression, leading to excellent inhibition of both primary tumor growth and lung metastasis.
Collapse
Affiliation(s)
- Yue Gao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Aiyu Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yanying Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Honghua Guo
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201600, China
| | - Liangyu He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Kangan Li
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201600, China
| | - Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of NASB, Akademicheskaja 27, 220072 Minsk, Belarus
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
8
|
Jia X, Wang Y, Qiao Y, Jiang X, Li J. Nanomaterial-based regulation of redox metabolism for enhancing cancer therapy. Chem Soc Rev 2024; 53:11590-11656. [PMID: 39431683 DOI: 10.1039/d4cs00404c] [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: 10/22/2024]
Abstract
Altered redox metabolism is one of the hallmarks of tumor cells, which not only contributes to tumor proliferation, metastasis, and immune evasion, but also has great relevance to therapeutic resistance. Therefore, regulation of redox metabolism of tumor cells has been proposed as an attractive therapeutic strategy to inhibit tumor growth and reverse therapeutic resistance. In this respect, nanomedicines have exhibited significant therapeutic advantages as intensively reported in recent studies. In this review, we would like to summarize the latest advances in nanomaterial-assisted strategies for redox metabolic regulation therapy, with a focus on the regulation of redox metabolism-related metabolite levels, enzyme activity, and signaling pathways. In the end, future expectations and challenges of such emerging strategies have been discussed, hoping to enlighten and promote their further development for meeting the various demands of advanced cancer therapies. It is highly expected that these therapeutic strategies based on redox metabolism regulation will play a more important role in the field of nanomedicine.
Collapse
Affiliation(s)
- Xiaodan Jia
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Wang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Qiao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xiue Jiang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Jinghong Li
- Beijing Institute of Life Science and Technology, Beijing 102206, P. R. China
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P. R. China.
| |
Collapse
|
9
|
Wang F, Liu Z, Liu Y, Zhang J, Xu W, Liu B, Sun Z, Chu H. A Spatiotemporally Controlled Gene-Regulation Strategy for Combined Tumor Therapy Based on Upconversion Hybrid Nanosystem. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405640. [PMID: 39207039 PMCID: PMC11515897 DOI: 10.1002/advs.202405640] [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: 05/23/2024] [Revised: 08/09/2024] [Indexed: 09/04/2024]
Abstract
The lack of precise spatiotemporal gene modulation and therapy impedes progress in medical applications. Herein, a 980 nm near-infrared (NIR) light-controlled nanoplatform, namely URMT, is developed, which can allow spatiotemporally controlled photodynamic therapy and trigger the enzyme-activated gene expression regulation in tumors. URMT is constructed by engineering an enzyme-activatable antisense oligonucleotide, which combined with an upconversion nanoparticle (UCNP)-based photodynamic nanosystem, followed by the surface functionalization of triphenylphosphine (TPP), a mitochondria-targeting ligand. URMT allows for the 980 nm NIR light-activated generation of reactive oxygen species, which can induce the translocation of a DNA repair enzyme (namely apurinic/apyrimidinic endonuclease 1, APE1) from the nucleus to mitochondria. APE1 can recognize the basic apurinic/apyrimidinic (AP) sites in DNA double-strands and perform cleavage, thereby releasing the functional single-strands for gene regulation. Overall, an augmented antitumor effect is observed due to NIR light-controlled mitochondrial damage and enzyme-activated gene regulation. Altogether, the approach reported in this study offers high spatiotemporal precision and shows the potential to achieve precise and specific gene regulation for targeted tumor treatment.
Collapse
Affiliation(s)
- Fang Wang
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Zechao Liu
- College of ScienceMinzu University of China27 Zhongguancun South AvenueBeijing100081China
| | - Yuechen Liu
- College of ScienceMinzu University of China27 Zhongguancun South AvenueBeijing100081China
| | - Jiayi Zhang
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Weizhe Xu
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Bei Liu
- College of ScienceMinzu University of China27 Zhongguancun South AvenueBeijing100081China
| | - Zhaogang Sun
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Hongqian Chu
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| |
Collapse
|
10
|
Wang J, Liu Y, Cui T, Yang H, Lin L. Current progress in the regulation of endogenous molecules for enhanced chemodynamic therapy. Chem Sci 2024; 15:9915-9926. [PMID: 38966366 PMCID: PMC11220580 DOI: 10.1039/d4sc02129k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/05/2024] [Indexed: 07/06/2024] Open
Abstract
Chemodynamic therapy (CDT) is a potential cancer treatment strategy, which relies on Fenton chemistry to transform hydrogen peroxide (H2O2) into highly cytotoxic reactive oxygen species (ROS) for tumor growth suppression. Although overproduced H2O2 in cancerous tissues makes CDT a feasible and specific tumor therapeutic modality, the treatment outcomes of traditional chemodynamic agents still fall short of expectations. Reprogramming cellular metabolism is one of the hallmarks of tumors, which not only supports unrestricted proliferative demands in cancer cells, but also mediates the resistance of tumor cells against many antitumor modalities. Recent discoveries have revealed that various cellular metabolites including H2O2, iron, lactate, glutathione, and lipids have distinct effects on CDT efficiency. In this perspective, we intend to provide a comprehensive summary of how different endogenous molecules impact Fenton chemistry for a deep understanding of mechanisms underlying endogenous regulation-enhanced CDT. Moreover, we point out the current challenges and offer our outlook on the future research directions in this field. We anticipate that exploring CDT through manipulating metabolism will yield significant advancements in tumor treatment.
Collapse
Affiliation(s)
- Jun Wang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| | - Yina Liu
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| | - Tingting Cui
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore Singapore 119074 Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore Singapore 117597 Singapore
| | - Huanghao Yang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| | - Lisen Lin
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| |
Collapse
|
11
|
Liu C, Xu X, Chen Y, Yin M, Mäkilä E, Zhou W, Su W, Zhang H. Metabolism-Regulating Nanozyme System for Advanced Nanocatalytic Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307794. [PMID: 38168483 DOI: 10.1002/smll.202307794] [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: 09/07/2023] [Revised: 12/17/2023] [Indexed: 01/05/2024]
Abstract
Nanocatalytic therapy, an emerging approach in cancer treatment, utilizes nanomaterials to initiate enzyme-mimetic catalytic reactions within tumors, inducing tumor-suppressive effects. However, the targeted and selective catalysis within tumor cells is challenging yet critical for minimizing the adverse effects. The distinctive reliance of tumor cells on glycolysis generates abundant lactate, influencing the tumor's pH, which can be manipulated to selectively activate nanozymatic catalysis. Herein, small interfering ribonucleic acid (siRNA) targeting lactate transporter-mediated efflux is encapsulated within the iron-based metal-organic framework (FeMOF) and specifically delivered to tumor cells through cell membrane coating. This approach traps lactate within the cell, swiftly acidifying the tumor cytoplasm and creating an environment for boosting the catalysis of the FeMOF nanozyme. The nanozyme generates hydroxyl radical (·OH) in the reversed acidic environment, using endogenous hydrogen peroxide (H2O2) produced by mitochondria as a substrate. The induced cytoplasmic acidification disrupts calcium homeostasis, leading to mitochondrial calcium overload, resulting in mitochondrial dysfunction and subsequent tumor cell death. Additionally, the tumor microenvironment is also remodeled, inhibiting migration and invasion, thus preventing metastasis. This groundbreaking strategy combines metabolic regulation with nanozyme catalysis in a toxic drug-free approach for tumor treatment, holding promise for future clinical applications.
Collapse
Affiliation(s)
- Chang Liu
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland
| | - Xiaoyu Xu
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200031, China
| | - Yongyang Chen
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Miao Yin
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Ermei Mäkilä
- Industrial Physics Laboratory, Department of Physics and Astronomy, University of Turku, Turku, 20014, Finland
| | - Wenhui Zhou
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | - Wenmei Su
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Hongbo Zhang
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| |
Collapse
|
12
|
Abdellatif AAH, Bouazzaoui A, Tawfeek HM, Younis MA. MCT4 knockdown by tumor microenvironment-responsive nanoparticles remodels the cytokine profile and eradicates aggressive breast cancer cells. Colloids Surf B Biointerfaces 2024; 238:113930. [PMID: 38692174 DOI: 10.1016/j.colsurfb.2024.113930] [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: 02/08/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Breast cancer is a wide-spread threat to the women's health. The drawbacks of conventional treatments necessitate the development of alternative strategies, where gene therapy has regained hope in achieving an efficient eradication of aggressive tumors. Monocarboxylate transporter 4 (MCT4) plays pivotal roles in the growth and survival of various tumors, which offers a promising target for treatment. In the present study, pH-responsive lipid nanoparticles (LNPs) based on the ionizable lipid,1,2-dioleoyl-3-dimethylammonium propane (DODAP), were designed for the delivery of siRNA targeting MCT4 gene to the breast cancer cells. Following multiple steps of characterization and optimization, the anticancer activities of the LNPs were assessed against an aggressive breast cancer cell line, 4T1, in comparison with a normal cell line, LX-2. The selection of the helper phospholipid to be incorporated into the LNPs had a dramatic impact on their gene delivery performance. The optimized LNPs enabled a powerful MCT4 silencing by ∼90 % at low siRNA concentrations, with a subsequent ∼80 % cytotoxicity to 4T1 cells. Meanwhile, the LNPs demonstrated a 5-fold higher affinity to the breast cancer cells versus the normal cells, in which they had a minimum effect. Moreover, the MCT4 knockdown by the treatment remodeled the cytokine profile in 4T1 cells, as evidenced by 90 % and ∼64 % reduction in the levels of TNF-α and IL-6; respectively. The findings of this study are promising for potential clinical applications. Furthermore, the simple and scalable delivery vector developed herein can serve as a breast cancer-targeting platform for the delivery of other RNA therapeutics.
Collapse
Affiliation(s)
- Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraidah 51452, Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Abdellatif Bouazzaoui
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; Science and Technology Unit, Umm Al-Qura University, Makkah 21955, Saudi Arabia; Department of Internal Medicine III (Haematology and Internal Oncology), University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg 93053, Germany
| | - Hesham M Tawfeek
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Mahmoud A Younis
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| |
Collapse
|
13
|
Lei J, Qi S, Yu X, Gao X, Yang K, Zhang X, Cheng M, Bai B, Feng Y, Lu M, Wang Y, Li H, Yu G. Development of Mannosylated Lipid Nanoparticles for mRNA Cancer Vaccine with High Antigen Presentation Efficiency and Immunomodulatory Capability. Angew Chem Int Ed Engl 2024; 63:e202318515. [PMID: 38320193 DOI: 10.1002/anie.202318515] [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: 12/03/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/08/2024]
Abstract
Insufficient accumulation of lipid nanoparticles (LNPs)-based mRNA vaccines in antigen presenting cells remains a key barrier to eliciting potent antitumor immune responses. Herein, we develop dendritic cells (DCs) targeting LNPs by taking advantage of mannose receptor-mediated endocytosis. Efficient delivery of mRNA to DCs is achieved in vitro and in vivo utilizing the sweet LNPs (STLNPs-Man). Intramuscular injection of mRNA vaccine (STLNPs-Man@mRNAOVA ) results in a four-fold higher uptake by DCs in comparison with commercially used LNPs. Benefiting from its DCs targeting ability, STLNPs-Man@mRNAOVA significantly promotes the antitumor performances, showing a comparable therapeutic efficacy by using one-fifth of the injection dosage as the vaccine prepared from normal LNPs, thus remarkably avoiding the side effects brought by conventional mRNA vaccines. More intriguingly, STLNPs-Man@mRNAOVA exhibits the ability to downregulate the expression of cytotoxic T-lymphocyte-associated protein 4 on T cells due to the blockade of CD206/CD45 axis, showing brilliant potentials in promoting antitumor efficacy combined with immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Jiaqi Lei
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Shaolong Qi
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Xinyang Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Xiaomin Gao
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Kai Yang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Xueyan Zhang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Meiqi Cheng
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Bing Bai
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Yunxuan Feng
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Meixin Lu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Yangfan Wang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Hongjian Li
- School of Medicine, Tsinghua University, 100084, Beijing, P. R. China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
- School of Medicine, Tsinghua University, 100084, Beijing, P. R. China
| |
Collapse
|
14
|
Deng W, Shang H, Tong Y, Liu X, Huang Q, He Y, Wu J, Ba X, Chen Z, Chen Y, Tang K. The application of nanoparticles-based ferroptosis, pyroptosis and autophagy in cancer immunotherapy. J Nanobiotechnology 2024; 22:97. [PMID: 38454419 PMCID: PMC10921615 DOI: 10.1186/s12951-024-02297-8] [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: 04/15/2023] [Accepted: 01/02/2024] [Indexed: 03/09/2024] Open
Abstract
Immune checkpoint blockers (ICBs) have been applied for cancer therapy and achieved great success in the field of cancer immunotherapy. Nevertheless, the broad application of ICBs is limited by the low response rate. To address this issue, increasing studies have found that the induction of immunogenic cell death (ICD) in tumor cells is becoming an emerging therapeutic strategy in cancer treatment, not only straightly killing tumor cells but also enhancing dying cells immunogenicity and activating antitumor immunity. ICD is a generic term representing different cell death modes containing ferroptosis, pyroptosis, autophagy and apoptosis. Traditional chemotherapeutic agents usually inhibit tumor growth based on the apoptotic ICD, but most tumor cells are resistant to the apoptosis. Thus, the induction of non-apoptotic ICD is considered to be a more efficient approach for cancer therapy. In addition, due to the ineffective localization of ICD inducers, various types of nanomaterials have been being developed to achieve targeted delivery of therapeutic agents and improved immunotherapeutic efficiency. In this review, we briefly outline molecular mechanisms of ferroptosis, pyroptosis and autophagy, as well as their reciprocal interactions with antitumor immunity, and then summarize the current progress of ICD-induced nanoparticles based on different strategies and illustrate their applications in the cancer therapy.
Collapse
Affiliation(s)
- Wen Deng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuan Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
15
|
Xu L, Cao Y, Xu Y, Li R, Xu X. Redox-Responsive Polymeric Nanoparticle for Nucleic Acid Delivery and Cancer Therapy: Progress, Opportunities, and Challenges. Macromol Biosci 2024; 24:e2300238. [PMID: 37573033 DOI: 10.1002/mabi.202300238] [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: 05/25/2023] [Revised: 07/25/2023] [Indexed: 08/14/2023]
Abstract
Cancer development and progression of cancer are closely associated with the activation of oncogenes and loss of tumor suppressor genes. Nucleic acid drugs (e.g., siRNA, mRNA, and DNA) are widely used for cancer therapy due to their specific ability to regulate the expression of any cancer-associated genes. However, nucleic acid drugs are negatively charged biomacromolecules that are susceptible to serum nucleases and cannot cross cell membrane. Therefore, specific delivery tools are required to facilitate the intracellular delivery of nucleic acid drugs. In the past few decades, a variety of nanoparticles (NPs) are designed and developed for nucleic acid delivery and cancer therapy. In particular, the polymeric NPs in response to the abnormal redox status in cancer cells have garnered much more attention as their potential in redox-triggered nanostructure dissociation and rapid intracellular release of nucleic acid drugs. In this review, the important genes or signaling pathways regulating the abnormal redox status in cancer cells are briefly introduced and the recent development of redox-responsive NPs for nucleic acid delivery and cancer therapy is systemically summarized. The future development of NPs-mediated nucleic acid delivery and their challenges in clinical translation are also discussed.
Collapse
Affiliation(s)
- Lei Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Yuan Cao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Ya Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Rong Li
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| |
Collapse
|
16
|
Cheng Q, Shi X, Li Q, Wang L, Wang Z. Current Advances on Nanomaterials Interfering with Lactate Metabolism for Tumor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305662. [PMID: 37941489 PMCID: PMC10797484 DOI: 10.1002/advs.202305662] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/15/2023] [Indexed: 11/10/2023]
Abstract
Increasing numbers of studies have shown that tumor cells prefer fermentative glycolysis over oxidative phosphorylation to provide a vast amount of energy for fast proliferation even under oxygen-sufficient conditions. This metabolic alteration not only favors tumor cell progression and metastasis but also increases lactate accumulation in solid tumors. In addition to serving as a byproduct of glycolytic tumor cells, lactate also plays a central role in the construction of acidic and immunosuppressive tumor microenvironment, resulting in therapeutic tolerance. Recently, targeted drug delivery and inherent therapeutic properties of nanomaterials have attracted great attention, and research on modulating lactate metabolism based on nanomaterials to enhance antitumor therapy has exploded. In this review, the advanced tumor therapy strategies based on nanomaterials that interfere with lactate metabolism are discussed, including inhibiting lactate anabolism, promoting lactate catabolism, and disrupting the "lactate shuttle". Furthermore, recent advances in combining lactate metabolism modulation with other therapies, including chemotherapy, immunotherapy, photothermal therapy, and reactive oxygen species-related therapies, etc., which have achieved cooperatively enhanced therapeutic outcomes, are summarized. Finally, foreseeable challenges and prospective developments are also reviewed for the future development of this field.
Collapse
Affiliation(s)
- Qian Cheng
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Xiao‐Lei Shi
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Qi‐Lin Li
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Lin Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Zheng Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| |
Collapse
|
17
|
Fateh ST, Fateh ST, Salehi-Najafabadi A, Aref AR. Commercial and regulatory challenges in cancer nanomedicine. FUNCTIONALIZED NANOMATERIALS FOR CANCER RESEARCH 2024:579-601. [DOI: 10.1016/b978-0-443-15518-5.00009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
18
|
Jia S, Ke S, Tu L, Chen S, Luo B, Xiong Y, Li Y, Wang P, Ye S. Glutathione/pH-responsive copper-based nanoplatform for amplified chemodynamic therapy through synergistic cycling regeneration of reactive oxygen species and dual glutathione depletion. J Colloid Interface Sci 2023; 652:329-340. [PMID: 37597414 DOI: 10.1016/j.jcis.2023.08.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/18/2023] [Accepted: 08/07/2023] [Indexed: 08/21/2023]
Abstract
The rapid scavenging of reactive oxygen species (ROS) by glutathione (GSH) and insufficient endogenous hydrogen peroxide (H2O2) in tumor cells are the major factors greatly restricting the efficacy of chemodynamic therapy (CDT). Herein, we developed a tumor microenvironment (TME)-responsive Cu-based metal-mesoporous organosilica nanoplatform integrating vitamin k3 (VK3), which could deplete GSH and specifically regenerate H2O2 for amplified CDT of cancer. Once the CuO@MON-PEG/VK3 nanoparticles entered into the tumor cells through enhanced permeability and retention (EPR) effect, the organosilicon shell and CuO core would be successively degraded upon the triggering of GSH and endo/lysosomal acidity. Subsequently, the enriched tetrasulfide bridges and released Cu2+ could consume GSH substantially, thus triggering Fenton-like reaction for CDT. Furthermore, the released VK3 could be catalyzed by the highly expressed quinone oxidoreductase-1 (NQO1) inside tumor cells to generate sufficient H2O2 through a "reversible" redox cycle, which in turn promoted Cu+-mediated Fenton-like reaction. Both in vitro and in vivo studies demonstrated that this nanoplatform could achieve synergistic CDT against tumor through synergistic cycling regeneration of ROS and dual GSH exhaustion with excellent biosafety. Our finding highlight the promising potential of CuO@MON-PEG/VK3 nanoplatform with multiple oxidative stress amplification for highly efficient tumor therapy.
Collapse
Affiliation(s)
- Sihan Jia
- Department of Biomaterials, The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, College of Materials, Xiamen University, Xiamen 361005, PR China
| | - Sunkui Ke
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, Xiamen 361004, PR China
| | - Li Tu
- Department of Biomaterials, The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, College of Materials, Xiamen University, Xiamen 361005, PR China
| | - Shengqiang Chen
- Department of Biomaterials, The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, College of Materials, Xiamen University, Xiamen 361005, PR China
| | - Bingkun Luo
- Department of Biomaterials, The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, College of Materials, Xiamen University, Xiamen 361005, PR China
| | - Yeqi Xiong
- Department of Biomaterials, The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, College of Materials, Xiamen University, Xiamen 361005, PR China
| | - Yang Li
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, PR China; Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, PR China.
| | - Peiyuan Wang
- Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, PR China.
| | - Shefang Ye
- Department of Biomaterials, The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, College of Materials, Xiamen University, Xiamen 361005, PR China.
| |
Collapse
|
19
|
Zhang J, Chen B, Gan C, Sun H, Zhang J, Feng L. A Comprehensive Review of Small Interfering RNAs (siRNAs): Mechanism, Therapeutic Targets, and Delivery Strategies for Cancer Therapy. Int J Nanomedicine 2023; 18:7605-7635. [PMID: 38106451 PMCID: PMC10725753 DOI: 10.2147/ijn.s436038] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/29/2023] [Indexed: 12/19/2023] Open
Abstract
Small interfering RNA (siRNA) delivery by nanocarriers has been identified as a promising strategy in the study and treatment of cancer. Short nucleotide sequences are synthesized exogenously to create siRNA, which triggers RNA interference (RNAi) in cells and silences target gene expression in a sequence-specific way. As a nucleic acid-based medicine that has gained popularity recently, siRNA exhibits novel potential for the treatment of cancer. However, there are still many obstacles to overcome before clinical siRNA delivery devices can be developed. In this review, we discuss prospective targets for siRNA drug design, explain siRNA drug properties and benefits, and give an overview of the current clinical siRNA therapeutics for the treatment of cancer. Additionally, we introduce the siRNA chemical modifications and delivery systems that are clinically sophisticated and classify bioresponsive materials for siRNA release in a methodical manner. This review will serve as a reference for researchers in developing more precise and efficient targeted delivery systems, promoting ongoing advances in clinical applications.
Collapse
Affiliation(s)
- Jiaying Zhang
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, People’s Republic of China
| | - Bo Chen
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, People’s Republic of China
| | - Chunyuan Gan
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, People’s Republic of China
| | - Hongyan Sun
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, People’s Republic of China
| | - Jiaxin Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
- Institute of Liver Diseases, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Lin Feng
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, People’s Republic of China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, People’s Republic of China
| |
Collapse
|
20
|
Peng C, Yu Z, Wu W, Li J, Wang E. CuFe Layered Double Hydroxide as Self-Cascade Nanoreactor for Efficient Antibacterial Therapy. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 38047886 DOI: 10.1021/acsami.3c11757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Nanozyme-induced reactive oxygen species (ROS)-dependent catalytic therapy has been developed into a powerful strategy against bacterial wound infections. However, the limited endogenous supply or instability of H2O2, the reliance on external stimuli for the generation of ROS, and the highly expressed glutathione (GSH) level make it a challenge to achieve high-performance therapeutic efficiency. In this work, a facile therapeutic strategy against bacterial infections with pristine CuFe layered double hydroxide (LDH) as the self-cascade nanoreactor is proposed without modification or additional energy input. CuFe LDH with an oxidase-like feature can catalyze the generation of multiple ROS, such as 1O2, ·O2-, and H2O2. And the self-generated H2O2 in the cascade nanoreactor could be further in situ transformed to ·OH owing to the peroxidase-like activity. As a result, the cell membrane of bacteria is destroyed, leading to death. Furthermore, its ultrahigh enzyme-like activity of CuFe LDH could effectively promote the breakdown of the biofilm structure. Additionally, the Cu2+-mediated GSH exhaustion of CuFe LDH further avoids the consumption of oxidized ROS and thereby significantly improves the sterilization effect. Finally, the as-prepared CuFe LDH with negligible side effects on normal tissues can be successfully used to eliminate the methicillin-resistant Staphylococcus aureus-infected wounds and accelerate their healing in the mouse model, which paves a new avenue as an antibacterial agent for clinical anti-infective treatment.
Collapse
Affiliation(s)
- Chao Peng
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Zhixuan Yu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Wenting Wu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jing Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Erkang Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
21
|
Mi J, Cui D, Zhang Z, Mu G, Shi Y. NIR-II femtosecond laser ignites MXene as photoacoustic bomb for continuous high-precision tumor blasting. NANOSCALE 2023; 15:16539-16551. [PMID: 37791688 DOI: 10.1039/d3nr03665k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Recently, photoacoustic (PA) cavitation-mediated therapy has become the focus of research owing to its advantage of inhibiting drug or radiation resistance; however, its application is limited because it relies on nanodroplets with one-time action. Herein, we demonstrate a femtosecond-laser-pumped ultrafast PA cavitation technique for highly efficient shockwave theranostics using niobium carbide (Nb2C) coated with polyvinylpyrrolidone-40000 (PVP), producing sustainable PA cavitation with non-phase-change nanoprobes, which effectively gets rid of the dependence on nanodroplets, guaranteeing multiple treatments. Under femtosecond (fs) laser irradiation, given that the thermal confinement regime could be well satisfied, the Nb2C-PVP nanosheets (NSs) were quickly heated, forming localized overheated nanospots with the temperature exceeding the phase-transition threshold of the surroundings, leading to precise cavitation and explosion at the tumor sites. The experiments at the cellular level showed the significant anti-tumor effects of this method. Notably, the mouse model experiments showed a relative tumor volume inhibition rate of more than 90%, demonstrating the high precision and good efficacy of the proposed anti-tumor method. This method provides a sustainable and highly effective strategy for PA theranostics, indicating its great potential for clinical applications.
Collapse
Affiliation(s)
- Jie Mi
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Dandan Cui
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Zhenhui Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Gen Mu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Yujiao Shi
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
22
|
Alkhathami AG, Sahib AS, Al Fayi MS, Fadhil AA, Jawad MA, Shafik SA, Sultan SJ, Almulla AF, Shen M. Glycolysis in human cancers: Emphasis circRNA/glycolysis axis and nanoparticles in glycolysis regulation in cancer therapy. ENVIRONMENTAL RESEARCH 2023; 234:116007. [PMID: 37119844 DOI: 10.1016/j.envres.2023.116007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 06/19/2023]
Abstract
The metabolism of cancer has been an interesting hallmark and metabolic reprogramming, especially the change from oxidative phosphorylation in mitochondria to glucose metabolism known as glycolysis occurs in cancer. The molecular profile of glycolysis, related molecular pathways and enzymes involved in this mechanism such as hexokinase have been fully understood. The glycolysis inhibition can significantly decrease tumorigenesis. On the other hand, circRNAs are new emerging non-coding RNA (ncRNA) molecules with potential biological functions and aberrant expression in cancer cells which have received high attention in recent years. CircRNAs have a unique covalently closed loop structure which makes them highly stable and reliable biomarkers in cancer. CircRNAs are regulators of molecular mechanisms including glycolysis. The enzymes involved in the glycolysis mechanism such as hexokinase are regulated by circRNAs to modulate tumor progression. Induction of glycolysis by circRNAs can significantly increase proliferation rate of cancer cells given access to energy and enhance metastasis. CircRNAs regulating glycolysis can influence drug resistance in cancers because of theirimpact on malignancy of tumor cells upon glycolysis induction. TRIM44, CDCA3, SKA2 and ROCK1 are among the downstream targets of circRNAs in regulating glycolysis in cancer. Additionally, microRNAs are key regulators of glycolysis mechanism in cancer cells and can affect related molecular pathways and enzymes. CircRNAs sponge miRNAs to regulate glycolysis as a main upstream mediator. Moreover, nanoparticles have been emerged as new tools in tumorigenesis suppression and in addition to drug and gene delivery, then mediate cancer immunotherapy and can be used for vaccine development. The nanoparticles can delivery circRNAs in cancer therapy and they are promising candidates in regulation of glycolysis, its suppression and inhibition of related pathways such as HIF-1α. The stimuli-responsive nanoparticles and ligand-functionalized ones have been developed for selective targeting of glycolysis and cancer cells, and mediating carcinogenesis inhibition.
Collapse
Affiliation(s)
- Ali G Alkhathami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Ameer S Sahib
- Department of Pharmacy, Al- Mustaqbal University College, 51001 Hilla, Iraq
| | - Majed Saad Al Fayi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Mohammed Abed Jawad
- Department of Medical Laboratories Technology, Al-Nisour University College, Iraq
| | - Sahar Ahmad Shafik
- Professor of Community Health Nursing, Faculty of Nursing, Fayum University, Egypt; College of Nursing, National University of Science and Technology, Iraq
| | | | - Abbas F Almulla
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Min Shen
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, China.
| |
Collapse
|
23
|
Zhang Y, Chen PH, Li B, Guo H, Zhu J, Dang Z, Lei S, Huang P, Lin J. Comprehensively Optimizing Fenton Reaction Factors for Antitumor Chemodynamic Therapy by Charge-Reversal Theranostics. ACS NANO 2023; 17:16743-16756. [PMID: 37616516 DOI: 10.1021/acsnano.3c03279] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Chemodynamic therapy (CDT) is a highly tumor-specific treatment, while its efficacy is compromised by the intratumoral Fenton reaction efficiency, which is determined by the following reaction factors, including the availability of Fenton ions (e.g., Fe2+), the amount of H2O2, and the degree of acidity. Synchronous optimization of these factors is a big challenge for efficient CDT. Herein, a strategy of comprehensively optimizing Fenton reaction factors was developed for traceable multistage augmented CDT by charge-reversal theranostics. The customized pH-responsive poly(ethylene)glycol-poly(β-amino esters) (PEG-PAE) micelle (PM) was prepared as the carrier. Glucose oxidase (GOx), Fe2+, and pH-responsive second near-infrared (NIR-II) LET-1052 probe were coloaded by PM to obtain the final theranostics. The activity of metastable Fe2+ remained by the unsaturated coordination with PEG-PAE. Then tumor accumulation and exposure of Fe2+ were achieved by charge-reversal cationization of PEG-PAE, which was further enhanced by a GOx catalysis-triggered pH decrease. Together with the abundant H2O2 generation and pH decrease through GOx catalysis, the limiting factors of the Fenton reaction were comprehensively optimized, achieving the enhanced CDT both in vitro and in vivo. These findings provide a strategy for comprehensively optimizing intratumoral Fenton reaction factors to overcome the intrinsic drawbacks of current CDT.
Collapse
Affiliation(s)
- Yajie Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Peng-Hang Chen
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Benhao Li
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Huishan Guo
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Junfei Zhu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Zechun Dang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Shan Lei
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| |
Collapse
|
24
|
Wu S, Xu L, He C, Wang P, Qin J, Guo F, Wang Y. Lactate Efflux Inhibition by Syrosingopine/LOD Co-Loaded Nanozyme for Synergetic Self-Replenishing Catalytic Cancer Therapy and Immune Microenvironment Remodeling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300686. [PMID: 37386815 PMCID: PMC10502866 DOI: 10.1002/advs.202300686] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/21/2023] [Indexed: 07/01/2023]
Abstract
An effective systemic mechanism regulates tumor development and progression; thus, a rational design in a one-stone-two-birds strategy is meant for cancer treatment. Herein, a hollow Fe3 O4 catalytic nanozyme carrier co-loading lactate oxidase (LOD) and a clinically-used hypotensor syrosingopine (Syr) are developed and delivered for synergetic cancer treatment by augmented self-replenishing nanocatalytic reaction, integrated starvation therapy, and reactivating anti-tumor immune microenvironment. The synergetic bio-effects of this nanoplatform stemmed from the effective inhibition of lactate efflux through blocking the monocarboxylate transporters MCT1/MCT4 functions by the loaded Syr as a trigger. Sustainable production of hydrogen peroxide by catalyzation of the increasingly residual intracellular lactic acid by the co-delivered LOD and intracellular acidification enabled the augmented self-replenishing nanocatalytic reaction. Large amounts of produced reactive oxygen species (ROS) damaged mitochondria to inhibit oxidative phosphorylation as the substituted energy supply upon the hampered glycolysis pathway of tumor cells. Meanwhile, remodeling anti-tumor immune microenvironment is implemented by pH gradient reversal, promoting the release of proinflammatory cytokines, restored effector T and NK cells, increased M1-polarize tumor-associated macrophages, and restriction of regulatory T cells. Thus, the biocompatible nanozyme platform achieved the synergy of chemodynamic/immuno/starvation therapies. This proof-of-concept study represents a promising candidate nanoplatform for synergetic cancer treatment.
Collapse
Affiliation(s)
- Shengming Wu
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering and Nano ScienceSchool of MedicineTongji UniversityShanghai200092P. R. China
| | - Lehua Xu
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering and Nano ScienceSchool of MedicineTongji UniversityShanghai200092P. R. China
| | - Chenlong He
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering and Nano ScienceSchool of MedicineTongji UniversityShanghai200092P. R. China
| | - Peng Wang
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering and Nano ScienceSchool of MedicineTongji UniversityShanghai200092P. R. China
| | - Jingwen Qin
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering and Nano ScienceSchool of MedicineTongji UniversityShanghai200092P. R. China
| | - Fangfang Guo
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering and Nano ScienceSchool of MedicineTongji UniversityShanghai200092P. R. China
| | - Yilong Wang
- The Institute for Translational NanomedicineShanghai East HospitalThe Institute for Biomedical Engineering and Nano ScienceSchool of MedicineTongji UniversityShanghai200092P. R. China
| |
Collapse
|
25
|
Koo S, Kim YG, Lee N, Hyeon T, Kim D. Inorganic nanoparticle agents for enhanced chemodynamic therapy of tumours. NANOSCALE 2023; 15:13498-13514. [PMID: 37578148 DOI: 10.1039/d3nr02000b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
With the recent interest in the role of oxidative species/radicals in diseases, inorganic nanomaterials with redox activities have been extensively investigated for their potential use in nanomedicine. While many studies focusing on relieving oxidative stress to prevent pathogenesis and to suppress the progression of diseases have shown considerable success, another approach for increasing oxidative stress using nanomaterials to kill malignant cells has suffered from low efficiency despite its wide applicability to various targets. Chemodynamic therapy (CDT) is an emerging technique that can resolve such a problem by exploiting the characteristic tumour microenvironment to achieve high selectivity. In this review, we summarize the recent strategies and underlying mechanisms that have been used to improve the CDT performance using inorganic nanoparticles. In addition to the design of CDT agents, the effects of contributing factors, such as the acidity and the levels of hydrogen peroxide and antioxidants in the tumour microenvironment, together with their modulation and application in combination therapy, are presented. The challenges lying ahead of future clinical translation of this rapidly advancing technology are also discussed.
Collapse
Affiliation(s)
- Sagang Koo
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.
| | - Young Geon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.
| | - Nohyun Lee
- School of Advanced Materials Engineering, Kookmin University, Seoul 02707, Republic of Korea.
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.
| | - Dokyoon Kim
- Department of Bionano Engineering, Hanyang University, Ansan 15588, Republic of Korea.
| |
Collapse
|
26
|
Pu Y, Ke H, Wu C, Xu S, Xiao Y, Han L, Lyv G, Li S. Superparamagnetic iron oxide nanoparticles target BxPC-3 cells and silence MUC4 for theranostics of pancreatic cancer. Biochim Biophys Acta Gen Subj 2023:130383. [PMID: 37236323 DOI: 10.1016/j.bbagen.2023.130383] [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: 11/20/2022] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023]
Abstract
PURPOSE Superparamagnetic iron oxide nanoparticles (SPION) are excellent magnetic resonance imaging (MRI) contrast agents. Mucin 4 (MUC4) acts as pancreatic cancer (PC) tumor antigen and influences PC progression. Small interfering RNAs (siRNAs) are used as a gene-silencing tool to treat a variety of diseases. METHODS We designed a therapeutic probe based on polyetherimide-superparamagnetic iron oxide nanoparticles (PEI-SPION) combined with siRNA nanoprobes (PEI-SPION-siRNA) to assess the contrast in MRI. The biocompatibility of the nanocomposite, and silencing of MUC4 were characterized and evaluated. RESULTS The prepared molecular probe had a particle size of 61.7 ± 18.5 nmand a surface of 46.7 ± 0.8mVand showed good biocompatibility in vitro and T2 relaxation efficiency. It can also load and protect siRNA. PEI-SPION-siRNA showed a good silencing effect on MUC4. CONCLUSION PEI-SPION-siRNA may be beneficial as a novel theranostic tool for PC.
Collapse
Affiliation(s)
- Yu Pu
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China; Department of Medical Imaging Key Laboratory of Sichuan Province, Affiliated Hospital of North Sichuan Medical College. No. 234, Fujiang Road, Shunqing District, Nanchong City 637000, People's Republic of China; Department of Medicine, Quanzhou Medical College, No. 2 Anji Road, Luojiang District, Quanzhou 362000, People's Republic of China
| | - Helin Ke
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Changqiang Wu
- Department of Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical College. No. 55, Dongshun Road, Gaoping District, Nanchong City 637100, People's Republic of China
| | - Shaodan Xu
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Yang Xiao
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Lina Han
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Guorong Lyv
- Department of Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical College. No. 55, Dongshun Road, Gaoping District, Nanchong City 637100, People's Republic of China.
| | - Shilin Li
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China.
| |
Collapse
|
27
|
Li T, Zhang Y, Zhu J, Zhang F, Xu A, Zhou T, Li Y, Liu M, Ke H, Yang T, Tang Y, Tao J, Miao L, Deng Y, Chen H. A pH-Activatable Copper-Biomineralized Proenzyme for Synergistic Chemodynamic/Chemo-Immunotherapy against Aggressive Cancers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210201. [PMID: 36573375 DOI: 10.1002/adma.202210201] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/19/2022] [Indexed: 06/18/2023]
Abstract
Artificial enzymes have demonstrated therapeutic benefits against diverse malignant tumors, yet their antitumor potencies are still severely compromised by non-selective catalysis, low atomic-utilization efficiency, and undesired off-target toxicity. Herein, it is reported that peroxidase-like biomineralized copper (II) carbonate hydroxide nanocrystals inside single albumin nanocages (CuCH-NCs) act as a pH-activatable proenzyme to achieve tumor-selective and synergistic chemodynamic/chemo-immunotherapy against aggressive triple-negative breast cancers (TNBCs). These CuCH-NCs show pH-sensitive Cu2+ release, which spontaneously undergoes glutathione (GSH)-mediated reduction into Cu+ species for catalyzing the evolution of H2 O2 into hydroxyl radicals (·OH) in a single-atom-like manner to cause chemodynamic cell injury, and simultaneously activates non-toxic disulfiram to cytotoxic complex for yielding selective chemotherapeutic damage via blocking cell proliferation and amplifying cell apoptosis. CuCH-NCs exhibit considerable tumor-targeting capacity with deep penetration depth, thus affording preferable efficacy against orthotopic breast tumors through synergistic chemodynamic/chemotherapy, together with good in vivo safety. Moreover, CuCH-NCs arouse distinct immunogenic cell death effect and upregulate PD-L1 expression upon disulfiram combination, and thus synergize with anti-PD-L1 antibody to activate adaptive and innate immunities, together with relieving immunosuppression, finally yielding potent antitumor efficacy against both primary and metastatic TNBCs. These results provide insights into smart and high-performance proenzymes for synergistic therapy against aggressive cancers.
Collapse
Affiliation(s)
- Ting Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Ying Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Jie Zhu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Fangrui Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - An'an Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Tian Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yaoqi Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Ming Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Hengte Ke
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Tao Yang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yong'an Tang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Jing Tao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, 215006, China
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Huabing Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, 215006, China
- State Key Laboratory of Radiation Medicine and Protection, and School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| |
Collapse
|
28
|
Gao H, Cao Z, Liu H, Chen L, Bai Y, Wu Q, Yu X, Wei W, Wang M. Multifunctional nanomedicines-enabled chemodynamic-synergized multimodal tumor therapy via Fenton and Fenton-like reactions. Theranostics 2023; 13:1974-2014. [PMID: 37064867 PMCID: PMC10091877 DOI: 10.7150/thno.80887] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/06/2023] [Indexed: 04/18/2023] Open
Abstract
Chemodynamic therapy (CDT) is well-known for using the tumor microenvironment to activate the Fenton reaction or Fenton-like reaction to generate strong oxidative hydroxyl radicals for tumor-specific treatment. It is highly selective and safe, without depth limitation of tissue penetration, and shows its potential as a new green therapeutic method with great clinical application. However, the catalytic efficiency of reagents involved in the Fenton reaction is severely affected by the inherent microenvironmental limitations of tumors and the strict Fenton reaction-dependent conditions. With the increasing application of nanotechnology in the medical field, combined therapies based on different types of functional nanomaterials have opened up new avenues for the development of next-generation CDT-enhanced system. This review will comprehensively exemplify representative results of combined therapies of CDT with other antitumor therapies such as chemotherapy, phototherapy, sonodynamic therapy, radiation therapy, magnetic hyperthermia therapy, immunotherapy, starvation therapy, gas therapy, gene therapy, oncosis therapy, or a combination thereof for improving antitumor efficiency from hundreds of the latest literature, introduce strategies such as the ingenious design of nanomedicines and tumor microenvironment regulations to enhance the combination therapy, and further summarize the challenges and future perspective of CDT-based multimodal anticancer therapy.
Collapse
Affiliation(s)
- Haiyan Gao
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, P. R. China
| | - Zhiping Cao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, 999077, China
| | - Huanhuan Liu
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, P. R. China
| | - Lijuan Chen
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, P. R. China
| | - Yan Bai
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, P. R. China
| | - Qingxia Wu
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, P. R. China
| | - Xuan Yu
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, P. R. China
| | - Wei Wei
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, P. R. China
| | - Meiyun Wang
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, P. R. China
- Laboratory of Brain Science and Brain-Like Intelligence Technology, Institute for Integrated Medical Science and Engineering, Henan Academy of Sciences, Zhengzhou, 450003, P. R. China
| |
Collapse
|
29
|
Zhang H, Wang Z, Gao T, Wang Z, Ren C, Liu J. An enzyme-instructed self-assembly system induces tumor acidosis via sequential-dual effect for cancer selective therapy. Acta Biomater 2023; 164:447-457. [PMID: 36996995 DOI: 10.1016/j.actbio.2023.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/06/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
The acidosis anti-tumor therapy, based on the altered energy metabolism pathway of tumor cells, has been proposed as an attractive method for cancer selective treatment. However, the strategy of inducing tumor acidosis by using a single drug to simultaneously inhibit both lactate efflux and consumption has not been reported yet. Herein, an in situ enzyme-instructed self-assembly (EISA) system was rationally fabricated to induce tumor acidosis apoptosis for cancer selective therapy. Depending on the sequential effect of the in situ EISA system, the targeted drug was successively distributed on the membrane and intracellular, inhibiting MCT4 mediated lactate efflux and mitochondrial tricarboxylic acid (TCA) cycle mediated lactate consumption, respectively. Through the dual obstruction of lactate metabolism to trigger tumor acidosis, the in situ EISA nanomedicine showed selective growth and migration inhibition against cancer cells. In addition, the nanomedicine also displayed a radio-sensitization effect in vitro due to causing the mitochondrial dysfunction, and exhibited a prominent synergistic chemo-radiotherapy anti-tumor performance in vivo. Accordingly, this work demonstrated that the in situ EISA system could endow the LND with sequential-dual effects to induce tumor acidosis, which may provide an enlightening strategy for anticancer drug delivery and cancer selective therapy. STATEMENT OF SIGNIFICANCE: With the help of the sequential effect of in situ EISA, the serial attack of LND against different targets was effectively realized to induce tumor acidosis and combined chemo-radiotherapy, implying the importance of the relationship between structure and function, which could offer a distinctive inspiration for future drug delivery system design and anti-tumor application.
Collapse
|
30
|
Recent advances in augmenting Fenton chemistry of nanoplatforms for enhanced chemodynamic therapy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.215004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
31
|
Yu H, Yan J, Li Z, Yang L, Ju F, Sun Y. Recent trends in emerging strategies for ferroptosis-based cancer therapy. NANOSCALE ADVANCES 2023; 5:1271-1290. [PMID: 36866253 PMCID: PMC9972547 DOI: 10.1039/d2na00719c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/31/2023] [Indexed: 06/18/2023]
Abstract
Ferroptosis, an iron-dependent mode of regulated cell death, is induced by lipid peroxidation, whose occurrence and execution are primarily controlled by metabolism of iron, lipids, amino acids and glutathione. In recent years, the fast-growing studies of ferroptosis in cancer have promoted its application in cancer therapy. So, this review focuses on the feasibility and characteristics of initiating ferroptosis for cancer therapy, as well as the main mechanism of ferroptosis. And various emerging strategies of cancer therapy based on ferroptosis are then highlighted to describe their design, mechanism of action, and anticancer applications. In addition ferroptosis in diverse cancer types is summarized, some considerations for the research of various preparations that can cause ferroptosis are introduced, and this emerging field is discussed in terms of its challenges and future development directions.
Collapse
Affiliation(s)
- Hongli Yu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266073 China
| | - Jianqin Yan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266073 China
| | - Zhipeng Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266073 China
| | | | - Fang Ju
- Department of Oncology, The Second Affiliated Hospital of Qingdao University Qingdao 266000 China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266073 China
| |
Collapse
|
32
|
Chen J, Zhu Y, Wu C, Shi J. Engineering lactate-modulating nanomedicines for cancer therapy. Chem Soc Rev 2023; 52:973-1000. [PMID: 36597879 DOI: 10.1039/d2cs00479h] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Lactate in tumors has long been considered "metabolic junk" derived from the glycolysis of cancer cells and utilized only as a biomarker of malignancy, but is presently believed to be a pivotal regulator of tumor development, maintenance and metastasis. Indeed, tumor lactate can be a "fuel" for energy supply and functions as a signaling molecule, which actively contributes to tumor progression, angiogenesis, immunosuppression, therapeutic resistance, etc., thus providing promising opportunities for cancer treatment. However, the current approaches for regulating lactate homeostasis with available agents are still challenging, which is mainly due to the short half-life, low bioavailability and poor specificity of these agents and their unsatisfactory therapeutic outcomes. In recent years, lactate modulation nanomedicines have emerged as a charming and efficient strategy for fighting cancer, which play important roles in optimizing the delivery of lactate-modulating agents for more precise and effective modulation and treatment. Integrating specific lactate-modulating functions in diverse therapeutic nanomedicines may overcome the intrinsic restrictions of different therapeutic modalities by remodeling the pathological microenvironment for achieving enhanced cancer therapy. In this review, the most recent advances in the engineering of functional nanomedicines that can modulate tumor lactate for cancer therapy are summarized and discussed, and the fundamental mechanisms by which lactate modulation benefits various therapeutics are elucidated. Finally, the challenges and perspectives of this emerging strategy in the anti-tumor field are highlighted.
Collapse
Affiliation(s)
- Jiajie Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China. .,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yufang Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China. .,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China. .,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China. .,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China.,Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200331, P. R. China
| |
Collapse
|
33
|
Rao Y, Fan T, Zhou L, Fang K, Sun Y, Hu X, Wang A, Li R, Zhu Z, Dong C, Shi S. A positive self-amplified H 2O 2 and acidity circulation for boosting CDT-PTT-starvation therapy. J Control Release 2023; 354:701-712. [PMID: 36690036 DOI: 10.1016/j.jconrel.2023.01.053] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023]
Abstract
The therapeutic application of chemodynamic therapy (CDT) is severely limited by the insufficient intracellular H2O2 and acidity in tumor. Herein, an acid-sensitive nanoplatform (ZIF67-ICG/TAM@GOx) to promote H2O2 and acidity enhancement through intracellular cyclic amplification for enhanced CDT is rationally designed. Notably, the acidic conditions of the tumor microenvironment (TME) can turn on the switch of the nanoplatform, setting free the loaded tamoxifen (TAM) and indocyanine green (ICG). The mitochondrial respiration inhibitor TAM and the superoxide dismutase-mimicking ZIF67 synergistically lead to an increase in the content of O2 and H2O2, accelerating the depletion of β-d-glucose by GOx to generate gluconate and H2O2. The gluconate in turn boosts the acidity to facilitate the collapse of nanoparticles, further significantly promoting the accumulation of intracellular H2O2 through a positive circulation. Consequently, the amplificated endogenous H2O2 is catalyzed by Co2+ to liberate hydroxyl radicals (•OH). Besides, ICG-mediated photothermal therapy (PTT) and GOx-induced starvation therapy along with CDT realize the synergistic cancer treatment. Importantly, in vitro and in vivo experiments verified that the nanoplatform performed superior specificity and excellent therapeutic responses. The smart nanoplatform overcomes H2O2 and acidity deficiency simultaneously for intensive CDT, providing new prospects for the development of biocompatible cancer synergistic therapy strategies.
Collapse
Affiliation(s)
- Yiming Rao
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Ting Fan
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Lulu Zhou
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Kang Fang
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Yanting Sun
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Xiaochun Hu
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Anqi Wang
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Ruihao Li
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Zhounan Zhu
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji University, Shanghai 200092, People's Republic of China
| | - Chunyan Dong
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji University, Shanghai 200092, People's Republic of China.
| | - Shuo Shi
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji University, Shanghai 200092, People's Republic of China.
| |
Collapse
|
34
|
Vangijzegem T, Lecomte V, Ternad I, Van Leuven L, Muller RN, Stanicki D, Laurent S. Superparamagnetic Iron Oxide Nanoparticles (SPION): From Fundamentals to State-of-the-Art Innovative Applications for Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15010236. [PMID: 36678868 PMCID: PMC9861355 DOI: 10.3390/pharmaceutics15010236] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/01/2023] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
Despite significant advances in cancer therapy over the years, its complex pathological process still represents a major health challenge when seeking effective treatment and improved healthcare. With the advent of nanotechnologies, nanomedicine-based cancer therapy has been widely explored as a promising technology able to handle the requirements of the clinical sector. Superparamagnetic iron oxide nanoparticles (SPION) have been at the forefront of nanotechnology development since the mid-1990s, thanks to their former role as contrast agents for magnetic resonance imaging. Though their use as MRI probes has been discontinued due to an unfavorable cost/benefit ratio, several innovative applications as therapeutic tools have prompted a renewal of interest. The unique characteristics of SPION, i.e., their magnetic properties enabling specific response when submitted to high frequency (magnetic hyperthermia) or low frequency (magneto-mechanical therapy) alternating magnetic field, and their ability to generate reactive oxygen species (either intrinsically or when activated using various stimuli), make them particularly adapted for cancer therapy. This review provides a comprehensive description of the fundamental aspects of SPION formulation and highlights various recent approaches regarding in vivo applications in the field of cancer therapy.
Collapse
Affiliation(s)
- Thomas Vangijzegem
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
- Correspondence: (T.V.); (S.L.)
| | - Valentin Lecomte
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
| | - Indiana Ternad
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
| | - Levy Van Leuven
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
| | - Robert N. Muller
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
- Center for Microscopy and Molecular Imaging (CMMI), Non-Ionizing Molecular Imaging Unit, 6041 Gosselies, Belgium
| | - Dimitri Stanicki
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
| | - Sophie Laurent
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, 7000 Mons, Belgium
- Center for Microscopy and Molecular Imaging (CMMI), Non-Ionizing Molecular Imaging Unit, 6041 Gosselies, Belgium
- Correspondence: (T.V.); (S.L.)
| |
Collapse
|
35
|
Shi Y, Zhang C, Liu C, Ma X, Liu Z. Image-Guided Precision Treatments. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1199:59-86. [PMID: 37460727 DOI: 10.1007/978-981-32-9902-3_4] [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: 07/20/2023]
Abstract
Chemotherapy, radiotherapy, and surgery are traditional cancer treatments, which usually produce unpredictable side effects and potential risks to normal healthy organs/tissues. Thus, safe and reliable treatment strategies are urgently required for maximized therapeutic efficiency to lesions and minimized risks to healthy regions. To this end, molecular imaging is responsible to undertake a specific targeting therapy. Besides that, the image guidance as a precision visualized approach for real-time in situ evaluations as well as an intraoperational navigation approach has earned attractive attention in the past decade. Along with the rapid development of multifunctional micro-/nanobiomaterials, versatile cutting-edge and advanced therapy strategies (e.g., thermal therapy, dynamic therapy, gas therapy, etc.) have been achieved and greatly contributed to the image-guided precision treatments in every aspect. Therefore, this chapter aims to discuss about both traditional and advanced cancer treatments and especially to elucidate the important roles that visualized medicine has been playing in the image-guided precision treatments.
Collapse
Affiliation(s)
- Yu Shi
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Chen Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Chenxi Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Xinyong Ma
- Division of Academic & Cultural Activities, Academic Divisions of the Chinese Academy of Sciences, Beijing, China
| | - Zhe Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| |
Collapse
|
36
|
Recent advances in targeted gene silencing and cancer therapy by nanoparticle-based delivery systems. Biomed Pharmacother 2023; 157:114065. [PMID: 36481408 DOI: 10.1016/j.biopha.2022.114065] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Nanomedicine has emerged as a promising platform for disease treatment and much progress has been achieved in the clinical translation for cancer treatment. Several types of nanomedicines have been approved for therapeutic application. However, many nanoparticles still suffer from challenges in the translation from bench to bedside. Currently, nanoparticle-based delivery systems have been developed to explore their functions in targeted gene silencing and cancer therapy. This review describes the research progress of different nano-carriers in targeted gene editing, and the recent progress in co-delivery of anticancer drugs and small ribonucleic acid. We also summarize the strategies for improving the specificity of carrier systems. Finally, we discuss the functions of targeted nano-carriers in overcoming chemotherapeutic drug resistance in cancer therapy. As research continues to advance, a better understanding of the safety including long-term toxicity, immunogenicity, and body metabolism may impel nanoparticle translation.
Collapse
|
37
|
Recent advances in multi-configurable nanomaterials for improved chemodynamic therapy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
38
|
Ojha A, Jaiswal S, Bharti P, Mishra SK. Nanoparticles and Nanomaterials-Based Recent Approaches in Upgraded Targeting and Management of Cancer: A Review. Cancers (Basel) 2022; 15:cancers15010162. [PMID: 36612158 PMCID: PMC9817889 DOI: 10.3390/cancers15010162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/29/2022] Open
Abstract
Along with the extensive improvement in tumor biology research and different therapeutic developments, cancer remains a dominant and deadly disease. Tumor heterogeneity, systemic toxicities, and drug resistance are major hurdles in cancer therapy. Chemotherapy, radiotherapy, phototherapy, and surgical therapy are some prominent areas of cancer treatment. During chemotherapy for cancer, chemotherapeutic agents are distributed all over the body and also damage normal cells. With advancements in nanotechnology, nanoparticles utilized in all major areas of cancer therapy offer the probability to advance drug solubility, and stability, extend drug half-lives in plasma, reduce off-target effects, and quintessence drugs at a target site. The present review compiles the use of different types of nanoparticles in frequently and recently applied therapeutics of cancer therapy. A recent area of cancer treatment includes cancer stem cell therapy, DNA/RNA-based immunomodulation therapy, alteration of the microenvironment, and cell membrane-mediated biomimetic approach. Biocompatibility and bioaccumulation of nanoparticles is the major impediment in nano-based therapy. More research is required to develop the next generation of nanotherapeutics with the incorporation of new molecular entities, such as kinase inhibitors, siRNA, mRNA, and gene editing. We assume that nanotherapeutics will dramatically improve patient survival, move the model of cancer treatment, and develop certainty in the foreseeable future.
Collapse
Affiliation(s)
- Anupama Ojha
- Department of Allied Health Science, Mahayogi Gorakhnath University, Gorakhpur 273007, India
| | - Sonali Jaiswal
- Department of Biotechnology, DDU Gorakhpur University, Gorakhpur 273009, India
| | - Priyanka Bharti
- Department of Biotechnology, DDU Gorakhpur University, Gorakhpur 273009, India
| | - Sarad Kumar Mishra
- Department of Biotechnology, DDU Gorakhpur University, Gorakhpur 273009, India
- Correspondence:
| |
Collapse
|
39
|
Wei X, Wang J, Liang M, Song M. Development of functional nanomedicines for tumor associated macrophages-focused cancer immunotherapy. Theranostics 2022; 12:7821-7852. [PMID: 36451865 PMCID: PMC9706587 DOI: 10.7150/thno.78572] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/23/2022] [Indexed: 12/03/2022] Open
Abstract
Clinical cancer immunotherapies are usually impeded by tumor immunosuppression driven by tumor associated macrophages (TAMs). Thus, TAMs can be considered as a promising therapeutic target for improved immunotherapy, and TAMs-focused molecular targeting agents have made ideal progress in clinical practice. Even so, most TAMs-targeting agents still cannot cover up their own shortcomings as free drugs. The emergence of multifunctional nanomaterials can expectedly endow these therapeutic cargoes with high solubility, favorable pharmacokinetic distribution, cell-specific delivery, and controlled release. Here, the underlying mechanisms of tumor immunosuppression caused by TAMs are first emphatically elucidated, and then the basic design of TAMs-focused immune-nanomedicines are discussed, mainly including diverse categories of nanomaterials, targeted and stimulus-responsive modifications, and TAM imaging in nanomedicines. A summary of current TAMs-targeting immunotherapeutic mechanisms based on functional nanomedicines for TAMs elimination and/or repolarization is further presented. Lastly, some severe challenges related to functional nanomedicines for TAMs-focused cancer immunotherapy are proposed, and some feasible perspectives on clinical translation of TAMs-associated anticancer immunonanomedicines are provided. It is hoped that, with rapid development of nanomedicine in cancer immunotherapy, TAMs-focused therapeutic strategies may be anticipated to become an emerging immunotherapeutic modality for future clinical cancer treatment.
Collapse
Affiliation(s)
- Xiao Wei
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, P. R. China
| | - Jing Wang
- Section of Molecular Dermatology, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - Min Liang
- Department of Thoracic and Cardiac Surgery, Affiliated Hospital of Chengdu University, Chengdu 610081, P. R. China
| | - Mingzhu Song
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, P. R. China
- Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
40
|
Zhang Y, Hu H, Deng X, Song Q, Xing X, Liu W, Zhang Y. Cascade-Enhanced Catalytic Nanocomposite with Glutathione Depletion and Respiration Inhibition for Effective Starving-Chemodynamic Therapy Against Hypoxic Tumor. Int J Nanomedicine 2022; 17:5491-5510. [DOI: 10.2147/ijn.s382750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/05/2022] [Indexed: 11/22/2022] Open
|
41
|
Advancements of Prussian blue-based nanoplatforms in biomedical fields: Progress and perspectives. J Control Release 2022; 351:752-778. [DOI: 10.1016/j.jconrel.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 12/07/2022]
|
42
|
Luo C, Xie Y, He M, Xia Y, Li Y, He L, Li J, Wang L, Han X, Zhang L, Yuan X, Wang Z, Liu Y, Tan W. Artificial Nucleobase-Directed Programmable Synthesis and Assembly of Amphiphilic Nucleic Acids as an All-in-One Platform for Cation-Free siRNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2022; 14:44019-44028. [PMID: 36149091 DOI: 10.1021/acsami.2c09406] [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: 06/16/2023]
Abstract
Efficient transport of nucleic acid therapeutics into targeted cells is the key step of genetic modulation in disease treatment. Nowadays, delivery systems strongly rely on cationic materials, but how to balance the trade-off between effectiveness and toxicity of these exogenous materials remains incredibly challenging. Here, we take inspiration from nucleic acid chemistry and introduce a new concept of amphiphilic nucleic acids (ANAs), as an all-in-one platform for cation-free nucleic acid delivery, by programmatically conjugating two different artifical nucleobases with sequence-independent activities. Specifically, the hydrophilic artificial nucleobases in ANAs act as both delivery vectors and therapeutic cargos for integrated benefits, while the hydrophobic nucleobases enable molecular self-assembly for improved stability and endosomal membrane oxidation for enhanced endosomal escape. By virtue of these merits, this platform is successfully used for short interference RNA (siRNA) delivery, which demonstrates a high siRNA loading capacity, rapid cellular uptake, and efficient endosomal escape, eliciting remarkable gene silencing and synergistic inhibitory effects on cancer cell proliferation and migration. This work is a case study in exploiting the basis of nucleic acid chemistry to afford new paradigms for advanced cancer theranostics.
Collapse
Affiliation(s)
- Can Luo
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Yuqi Xie
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Minze He
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Yinghao Xia
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Yazhou Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Lei He
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Jili Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Linlin Wang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Xiaoyan Han
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Lili Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Xi Yuan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Zhiqiang Wang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Yanlan Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hu-nan University, Changsha 410082, Hunan, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
- Institute of Molecular Medicine, Renji Hospital, Shanghai JiaoTong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
43
|
Mohammed DF, Madlool HA, Faris M, Shalan BH, Hasan HH, Azeez NF, Abbas FH. Harnessing inorganic nanomaterials for chemodynamic cancer therapy. Nanomedicine (Lond) 2022; 17:1891-1906. [PMID: 36647807 DOI: 10.2217/nnm-2022-0187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The most important aspect of chemodynamic therapy (CDT) is the harnessing of Fenton or Fenton-like chemistry for cancer therapy within the tumor microenvironment, which occurs because of the moderate acidity and overexpressed H2O2 in the tumor microenvironment. Hydroxyl radicals (•OH) produced within tumor cells via Fenton and Fenton-like reactions cause cancer cell death. Reactive oxygen species-mediated CDT demonstrates a desired anticancer impact without the need for external stimulation or the development of drug resistance. Cancer therapy based on CDT is known as a viable cancer therapy modality. This review discusses the most recent CDT advancements and provides some typical instances. As a result, potential methods for further improving CDT efficiency under the guidance of Fenton chemistry are offered.
Collapse
Affiliation(s)
- Dhelal F Mohammed
- Department of Pharmacy, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Hussein A Madlool
- Radiological Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Mohammed Faris
- Department of Dentistry, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Bashar Hadi Shalan
- Anesthesia Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Huda Hadi Hasan
- Department of Business Administration, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Nidaa F Azeez
- Department of Medical Physics, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Fatima Hashim Abbas
- Department of Medical Laboratory Techniques, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| |
Collapse
|
44
|
Xu Z, Li Q, Zhang C, Wang P, Xu X, Ran L, Zhang L, Tian G, Zhang G. Amorphous ferric oxide-coating selenium core-shell nanoparticles: a self-preservation Pt(IV) platform for multi-modal cancer therapies through hydrogen peroxide depletion-mediated anti-angiogenesis, apoptosis and ferroptosis. NANOSCALE 2022; 14:11600-11611. [PMID: 35861683 DOI: 10.1039/d2nr01837c] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A self-preservation Pt(IV) nanoplatform, amorphous ferric oxide-coating selenium core-shell nanoparticles (iAIO@NSe-Pt), was developed for H2O2 depletion-mediated tumor anti-angiogenesis, apoptosis, and ferroptosis. Upon entry into the blood, the ferric oxide shell effectively blocked the contact Pt(IV) prodrug with reduced molecules, then avoided the inactivation of the Pt(IV) prodrug and increased its accumulation in the tumor. After entering cancer cells, iAIO@NSe-Pt caused a series of cascade reactions: (1) AIO on the surface of iAIO@NSe-Pt quickly dissolved, released an abundance of Fe(II) because of the weakly acidic tumor microenvironment, and then catalyzed cellular H2O2 into highly toxic ˙OH, resulting in cellular H2O2 deficiency and cell ferroptosis. (2) The platinum(IV) prodrugs were exposed and quickly reduced to highly toxic Pt(II) by depleting GSH. This process inactivated GPX4, promoted ROS accumulation, and further accelerated ferroptosis. In addition, the generated Pt(II) quickly inhibited DNA replication, achieving effective apoptotic cell death. Meanwhile, Pt(II) inactivated SOD1, which blocked the synthesis of cellular H2O2 and accelerated ROS (superoxide anion radical) accumulation. (3) The deficiency of cellular H2O2 significantly inhibited the expression of vascular endothelial growth factor-A (VEGF-A), blocking tumor angiogenesis and then improving the anticancer effect. (4) After such a cascade reaction, the exposed NSe successively disrupted mitochondrial respiration and inhibited cancer angiogenesis, further inducing cancer cell death. Collectively, our functional and mechanical investigation suggested that iAIO@NSe-Pt exhibits excellent tumor targeting, biocompatibility and anti-tumor efficiency in vitro and in vivo, and provides a novel example of a self-preservation Pt(IV) nanoplatform for H2O2 depletion-mediated tumor anti-angiogenesis, apoptosis, and ferroptosis, showing great promise for future clinical use.
Collapse
Affiliation(s)
- Zhaowei Xu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, 264003, P. R. China.
| | - Qingdong Li
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, 264003, P. R. China.
| | - Caiyun Zhang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, 264003, P. R. China.
| | - Peng Wang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, 264003, P. R. China.
| | - Xiaotong Xu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, 264003, P. R. China.
| | - Lang Ran
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, 264003, P. R. China.
| | - Li Zhang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui 230022, P. R. China.
| | - Geng Tian
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, 264003, P. R. China.
| | - Guilong Zhang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, 264003, P. R. China.
| |
Collapse
|
45
|
Liu M, Zhang G. Amorphous Goethite as a Catalyst of Chemoselectivity Epoxidation of Alkenes by Hydrogen Peroxide. RUSS J GEN CHEM+ 2022. [DOI: 10.1134/s1070363222080230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
46
|
Hong Y, Tao Q, Liu YY, Wang Z, Wang H, Sun L. Copper peroxide coated upconversion nanoparticle modified with glucose oxidase for H 2O 2 self-supplying starvation-enhanced chemodynamic therapy in vitro. Dalton Trans 2022; 51:11325-11334. [PMID: 35838196 DOI: 10.1039/d2dt00163b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Chemodynamic therapy (CDT) can convert endogenous hydrogen peroxide (H2O2) to highly reactive hydroxyl radical (˙OH) through Fenton or Fenton-like reaction to kill tumor cells, which is a promising anticancer strategy. However, the limited H2O2 and overexpressed glutathione (GSH) in tumor cells make CDT ineffective. Here, an efficient nanocomposite, UCN@CuO2-GOx (UCCuG), was synthesized, realizing both starvation therapy and H2O2 self-supplying CDT in vitro. In this case, the glucose oxidase (GOx) of the nanocomposite could consume glucose for starvation therapy after the UCCuG nanocomposite entered tumor cells. In addition, the acidic environment of the lysosome triggered the release of Cu2+ and H2O2 by the decomposition of UCCu; then, Cu2+ was reduced to Cu+ by GSH in tumor cells; and finally, Cu+ catalyzed the released H2O2 to generate ˙OH for CDT. The in vitro experiments demonstrated starvation-enhanced CDT with remarkable results. Meanwhile, under 980 nm laser irradiation, the upconversion luminescence signal of UCN in the UCCuG nanocomposite was reduced due to the CuO2-GOx coating, while it gradually recovered after the UCCuG nanocomposite reacted with glucose and GSH under the tumor microenvironment (TME). Such a luminescent intensity recovery process is expected to monitor the TME-activated therapeutic effect in real time. This strategy may solve the problem of insufficient CDT efficacy caused by limited endogenous H2O2 and overexpressed GSH in tumor cells. This multifunctional nanocomposite demonstrates the promising application of starvation-enhanced CDT in tumor treatment.
Collapse
Affiliation(s)
- Yale Hong
- Department of Chemistry, College of Sciences, Shanghai University, Shanghai 200444, China. .,Research Center of Nano Science and Technology, College of Sciences, Shanghai University, Shanghai 200444, China
| | - Qinfeng Tao
- Research Center of Nano Science and Technology, College of Sciences, Shanghai University, Shanghai 200444, China
| | - Yuan-Yuan Liu
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| | - Zhuo Wang
- State Key Laboratory of Marine Resource Utilization in South China Sea & Special Glass Key Lab of Hainan Province, School of Information and Communication Engineering, Hainan University, Haikou 570228, China
| | - Haifang Wang
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| | - Lining Sun
- Department of Chemistry, College of Sciences, Shanghai University, Shanghai 200444, China. .,Research Center of Nano Science and Technology, College of Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|
47
|
Li J, Wei J, Gao Y, Zhao Q, Sun J, Ouyang J, NaNa. Peptide-assembled siRNA nanomicelles confine MnO -loaded silicages for synergistic chemical and gene-regulated cancer therapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
48
|
Chen W, Liu J, Zheng C, Bai Q, Gao Q, Zhang Y, Dong K, Lu T. Research Progress on Improving the Efficiency of CDT by Exacerbating Tumor Acidification. Int J Nanomedicine 2022; 17:2611-2628. [PMID: 35712639 PMCID: PMC9196673 DOI: 10.2147/ijn.s366187] [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: 03/12/2022] [Accepted: 05/16/2022] [Indexed: 12/21/2022] Open
Abstract
In recent years, chemodynamic therapy (CDT) has received extensive attention as a novel means of cancer treatment. The CDT agents can exert Fenton and Fenton-like reactions in the acidic tumor microenvironment (TME), converting hydrogen peroxide (H2O2) into highly toxic hydroxyl radicals (·OH). However, the pH of TME, as an essential factor in the Fenton reaction, does not catalyze the reaction effectively, hindering its efficiency, which poses a significant challenge for the future clinical application of CDT. Therefore, this paper reviews various strategies to enhance the antitumor properties of nanomaterials by modulating tumor acidity. Ultimately, the performance of CDT can be further improved by inducing strong oxidative stress to produce sufficient ·OH. In this paper, the various acidification pathways and proton pumps with potential acidification functions are mainly discussed, such as catalytic enzymes, exogenous acids, CAIX, MCT, NHE, NBCn1, etc. The problems, opportunities, and challenges of CDT in the cancer field are also discussed, thereby providing new insights for the design of nanomaterials and laying the foundation for their future clinical applications.
Collapse
Affiliation(s)
- Wenting Chen
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Jinxi Liu
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Caiyun Zheng
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Que Bai
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Qian Gao
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Yanni Zhang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Kai Dong
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710072, People's Republic of China
| | - Tingli Lu
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| |
Collapse
|
49
|
Jin X, Zhao H, Chao Z, Wang X, Zhang Q, Ju H, Liu Y. Self-assembled Cupric Oxide Nanoclusters for Highly efficient chemodynamic therapy. Chem Asian J 2022; 17:e202200296. [PMID: 35713338 DOI: 10.1002/asia.202200296] [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: 03/23/2022] [Revised: 06/04/2022] [Indexed: 11/11/2022]
Abstract
Chemodynamic therapy (CDT) based on Fenton and Fenton-like reactions induces cancer cell killing via in situ catalyzing H2 O2 and generating highly oxidative hydroxyl radicals (⋅OH) in tumor sites. Their application is not limited by tumor grown depth or hypoxic microenvironment. However, the reaction efficiency is still hampered due to the structure of catalytic agents and the requirement for low pH environment. Here, we design a porous CuO nanocluster (CuO NC) through self-assembly of oleylamine stabilized CuO NPs (OAm-CuO NPs), and functionalize it with folic acid (CuO NC-FA) for specific tumor cell targeting. It can catalyze H2 O2 with high efficiency in nearly neutral environment. Besides, the porous structure of CuO NC also helps the diffusion of H2 O2 to the interior of nanocluster to further improve Fenton-like reaction efficiency. The convenient synthesis of CuO NC-FA with good Fenton-like reaction efficiency at neutral environment demonstrates good chemodynamic therapy effect.
Collapse
Affiliation(s)
- Xinyu Jin
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R China
| | - Hongxia Zhao
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R China
| | - Zhicong Chao
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R China
| | - Xiaofeng Wang
- Department of Urology Affiliated Drum Tower Hospital, Medical School of Nanjing University Institute of Urology, Nanjing University, Nanjing, 210008, P. R. China
| | - Qing Zhang
- Department of Urology Affiliated Drum Tower Hospital, Medical School of Nanjing University Institute of Urology, Nanjing University, Nanjing, 210008, P. R. China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R China
| | - Ying Liu
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R China.,Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
50
|
Zheng Y, Han Y, Sun Q, Li Z. Harnessing anti-tumor and tumor-tropism functions of macrophages via nanotechnology for tumor immunotherapy. EXPLORATION (BEIJING, CHINA) 2022; 2:20210166. [PMID: 37323705 PMCID: PMC10190945 DOI: 10.1002/exp.20210166] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/10/2022] [Indexed: 06/15/2023]
Abstract
Reprogramming the immunosuppressive tumor microenvironment by modulating macrophages holds great promise in tumor immunotherapy. As a class of professional phagocytes and antigen-presenting cells in the innate immune system, macrophages can not only directly engulf and clear tumor cells, but also play roles in presenting tumor-specific antigen to initiate adaptive immunity. However, the tumor-associated macrophages (TAMs) usually display tumor-supportive M2 phenotype rather than anti-tumor M1 phenotype. They can support tumor cells to escape immunological surveillance, aggravate tumor progression, and impede tumor-specific T cell immunity. Although many TAMs-modulating agents have shown great success in therapy of multiple tumors, they face enormous challenges including poor tumor accumulation and off-target side effects. An alternative solution is the use of advanced nanostructures, which not only can deliver TAMs-modulating agents to augment therapeutic efficacy, but also can directly serve as modulators of TAMs. Another important strategy is the exploitation of macrophages and macrophage-derived components as tumor-targeting delivery vehicles. Herein, we summarize the recent advances in targeting and engineering macrophages for tumor immunotherapy, including (1) direct and indirect effects of macrophages on the augmentation of immunotherapy and (2) strategies for engineering macrophage-based drug carriers. The existing perspectives and challenges of macrophage-based tumor immunotherapies are also highlighted.
Collapse
Affiliation(s)
- Yanhui Zheng
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouChina
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouChina
| | - Qiao Sun
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouChina
| | - Zhen Li
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouChina
| |
Collapse
|