1
|
Gou R, Chen S, Lei Y, Wu P, Chen X, Zhang Q. Hypoxia Inhibitor Improves Iodine Uptake Disorder in Thyroid Cancer Through the hsa_circ_0023990/miR-448/DNMT1/NIS Axis. Cancer Sci 2025. [PMID: 40386902 DOI: 10.1111/cas.70102] [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: 01/13/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/20/2025] Open
Abstract
This research seeks to investigate how hypoxia inhibitors enhance iodine uptake in thyroid cancer cells. Clinical samples were gathered and assessed for hsa_circ_0023990, DNMT1, NIS, and their promoter methylation levels using RT-PCR, western blot, and methylation-specific PCR (MSP) techniques. The study involved examining the impact and mechanism of hsa_circ_0023990 on iodine uptake in differentiated thyroid carcinoma (DTC) cells through genetic manipulation. Luciferase reporter gene experiments were conducted to validate the interaction between hsa_circ_0023990, DNMT1, and miR-448. Xenograft tumors were established in nude mice for in vivo validation. The results showed that hsa_circ_0023990 was notably elevated in DTC and RAIR-DTC tissues. It was found that hsa_circ_0023990 could modulate NIS promoter methylation via the miR-448/DNMT1 signaling pathway, thereby influencing NIS expression. Hypoxia inhibitors effectively suppressed hsa_circ_0023990 expression in DTC cells, leading to increased NIS expression and enhanced iodine uptake. Subcutaneous transplantation experiments in animals further confirmed that hypoxia inhibitors could boost iodine absorption in tumor tissue and inhibit tumor growth through the hsa_circ_0023990/miR-448/DNMT1/NIS signaling axis. In conclusion, hypoxia inhibitors ameliorate iodine uptake dysfunction in thyroid cancer by acting on the hsa_circ_0023990/miR-448/DNMT1/NIS signaling pathway.
Collapse
Affiliation(s)
- Ruiqin Gou
- Department of Nuclear Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shiqi Chen
- Department of Nuclear Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yangyang Lei
- Department of Nuclear Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Pengqing Wu
- Department of Nuclear Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xuezhong Chen
- Department of Nuclear Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qing Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
2
|
Wang C, Zha YL, Wang H, Sun B, Qiang WG, Yuan Y, Shi HB, Hu WW. Carfilzomib promotes Iodine-125 seed radiation-induced apoptosis, paraptosis, and ferroptosis in esophageal squamous cell carcinoma by aggravating endoplasmic reticulum stress. Transl Oncol 2025; 57:102393. [PMID: 40315760 DOI: 10.1016/j.tranon.2025.102393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 02/09/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025] Open
Abstract
Iodine-125 (125I) seed brachytherapy has been applied to treat various malignant tumors such as esophageal cancer, however, radioresistance can reduce its efficacy. Endoplasmic reticulum stress (ERS) and subsequent unfolded protein response (UPR) is one of the core mechanisms of 125I seed radiation-induced cell death, thus aggravating ERS has been considered a promising sensitization strategy. Herein, we show that combination therapy of an irreversible proteasome inhibitor carfilzomib (CFZ) and 125I seed radiation displayed strong anti-tumor effect on esophageal squamous cell carcinoma (ESCC). Mechanistically, ERS and UPR regulated multiple cell death modalities induced by the combination therapy, including apoptosis, paraptosis, and ferroptosis. 125I seed radiation induced reactive oxygen species (ROS) production, DNA damage, p53 activation, and apoptosis. CFZ promoted ROS production, and augmented 125I seed radiation-induced apoptosis via the mitochondrial pathway, which was mediated by the UPR-C/EBP homologous protein (CHOP) pathway and was independent of the p53 pathway. CFZ enhanced 125I seed radiation-induced intracellular Ca2+ overload, protein ubiquitination, ERS, and UPR, consequently promoting paraptosis. 125I seed radiation induced accumulation of intracellular Fe2+ and lipid peroxides but upregulated the expression of ferroptosis inhibitors, SLC7A11 and glutathione peroxidase 4 (GPX4). The combination therapy promoted ferroptosis by enhancing the accumulation of intracellular Fe2+ and downregulating GPX4 expression. The mouse experiment demonstrated that CFZ can promote the efficacy of 125I seed radiation with good tolerance. Our findings suggest that combination therapy of 125I seed radiation and CFZ is associated with multiple cell death modalities and may serve as a promising therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Chao Wang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Yin-Lin Zha
- Department of Radiation Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Hao Wang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Bai Sun
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Wei-Guang Qiang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Ye Yuan
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Hong-Bing Shi
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China.
| | - Wen-Wei Hu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China.
| |
Collapse
|
3
|
Zhao Q, Han Y, Gong W, Cao Z, Chang H, Gong S, Li Q, Li M, Ma C, He L, Zhou H. Membrane-Anchoring and Oxygen-Generating Mediated Nanosonosensitizer for Optimizing Cancer Immunotherapy. Adv Healthc Mater 2025; 14:e2404849. [PMID: 40129017 DOI: 10.1002/adhm.202404849] [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: 12/03/2024] [Revised: 03/10/2025] [Indexed: 03/26/2025]
Abstract
Despite its antitumor promise, sonodynamic therapy (SDT)'s efficacy in immune activation requires enhancement, primarily due to the hypoxic tumor microenvironment (TME) and insufficient targeting of sonosensitizers to specific subcellular regions. Herein, we developed macrophage membrane (MM)-intermingled membrane fusogenic liposomes (MFL) to optimize sonoimmunotherapy that encapsulate catalase (CAT) within the core and incorporate the sonosensitizer chlorin e6 (Ce6) in the outer shell (CAT@MM-MFL-Ce6). The MM confers immune evasion properties and promotes nanoparticles' targeted accumulation in tumor tissue. The membrane fusion effect enables Ce6 to anchor onto cancer cell membrane and facilitates the direct delivery of CAT into the cytoplasm, bypassing endosomal degradation. Upon ultrasound stimulation, generated reactive oxygen species directly damage the plasma membrane, initiating the Caspase 3/Gasdermin E-mediated pyroptosis pathway. Concurrently, the encapsulated CAT efficiently decompose H₂O₂ in the cytoplasm, thus enhancing local oxygen levels in hypoxic tumors. Contributed by these effects, the combination of nanosonosensitizer-augmented SDT and immune checkpoint agent successfully reverse the immunosuppressive TME, driving a potent immune response that inhibits primary tumor growth, distant metastasis, and lung metastases in an orthotopic triple-negative breast cancer model. This study demonstrates the potential of a novel SDT-based combinatorial approach to modulate immune-cold TMEs, advancing proof-of-concept tumor therapeutics.
Collapse
Affiliation(s)
- Qing Zhao
- Ultrasound Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Surgeons' Hall, No.246. XuefuRoad, Nangang District, Harbin, Heilongjiang Province, 150086, P. R. China
| | - Ye Han
- In-Patient Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Surgeons' Hall, No.246. XuefuRoad, Nangang District, Harbin, Heilongjiang Province, 150086, P. R. China
| | - Wushuang Gong
- In-Patient Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Surgeons' Hall, No.246. XuefuRoad, Nangang District, Harbin, Heilongjiang Province, 150086, P. R. China
| | - Zhiyue Cao
- School of Medicine and Health, Key Laboratory of Microsystems and Microstructures Manufacturing, Harbin Institute of Technology, Harbin, 150001, P. R. China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, 450046, P. R. China
| | - Haonan Chang
- In-Patient Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Surgeons' Hall, No.246. XuefuRoad, Nangang District, Harbin, Heilongjiang Province, 150086, P. R. China
| | - Shaofan Gong
- In-Patient Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Surgeons' Hall, No.246. XuefuRoad, Nangang District, Harbin, Heilongjiang Province, 150086, P. R. China
| | - Qunying Li
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Mengmeng Li
- School of Medicine and Health, Key Laboratory of Microsystems and Microstructures Manufacturing, Harbin Institute of Technology, Harbin, 150001, P. R. China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, 450046, P. R. China
| | - Chao Ma
- In-Patient Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Surgeons' Hall, No.246. XuefuRoad, Nangang District, Harbin, Heilongjiang Province, 150086, P. R. China
| | - Liangcan He
- School of Medicine and Health, Key Laboratory of Microsystems and Microstructures Manufacturing, Harbin Institute of Technology, Harbin, 150001, P. R. China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, 450046, P. R. China
| | - Hang Zhou
- In-Patient Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Surgeons' Hall, No.246. XuefuRoad, Nangang District, Harbin, Heilongjiang Province, 150086, P. R. China
| |
Collapse
|
4
|
Song J, Kang F, Chang X, Wu J, Zhang P, Wang X, Wang F. Irradiation of 125I seeds blocks glycolysis in pancreatic cancer by inhibiting KLF5 m6A methylation through the suppression of RBM15. Exp Cell Res 2025; 448:114538. [PMID: 40147709 DOI: 10.1016/j.yexcr.2025.114538] [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: 02/09/2025] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 03/29/2025]
Abstract
This paper investigated whether 125I seed irradiation for pancreatic cancer treatment was mediated through the RBM15/KLF5 pathway in glycolysis. The study collected peripheral blood from pancreatic cancer patients, and detected the expression of RBM15 and KLF5 expression in the serum of pancreatic cancer patients before and after 125I seed irradiation. An in vitro study was conducted to investigate the effects 125I seed irradiation on the malignant behavior and glycolysis of pancreatic cancer cells. The underlying mechanisms were thoroughly examined through a series of logical experiments, including Western blot analysis, Dot-blot experiment, methylated RNA immunoprecipitation assay, and RNA pull down assay. A xenograft tumor model in nude mice was established and treated with 125I seed irradiation, which was employed to research the in vivo effect and mechanism of 125I seed irradiation for pancreatic cancer. The overexpressed RBM15 and KLF5 in serum of pancreatic cancer patients were reduced after 125I seed treatment. 125I seed treatment impaired pancreatic cancer cell proliferation and invasion; enhanced apoptosis; attenuated glycolysis; and reduced RBM15 and KLF5 expression. RBM15 overexpression partially reversed these influences of 125I seed treatment on pancreatic cancer cells. RBM15 was capable of increasing KLF5 expression, which might be achieved by promoting m6A methylation of KLF5. In vivo, 125I seed treatment blocked the growth of pancreatic cancer cells and decreased RBM15 and KLF5 expression in xenograft tumor, whereas RBM15 overexpression abolished these effects. 125I seed irradiation suppressed glycolysis in pancreatic cancer by inhibiting KLF5 m6A methylation through down-regulation of RBM15. This discovery established a solid foundation for the use of in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jianjun Song
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, China
| | - Fuping Kang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, China
| | - Xiaoning Chang
- Department of Rehabilitation Medicine, General Hospital of Ningxia Medical University, China
| | - Jing Wu
- Medical Experiment Center, General Hospital of Ningxia Medical University, China
| | - Peng Zhang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, China
| | - Xiaolong Wang
- School of Nursing, Zhengzhou Institute of Industrial Application Technology, China
| | - Feng Wang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, China.
| |
Collapse
|
5
|
Wang Z, Feng C, Lu S, Wang Y, Suo R, Jia K, Sun T, Mei J, Huang T, Bai X, Chen Q, Yao X, Wu B, Guo J, Lu J. Nanoscale CaO 2-Loaded Surface-Engineered Iodine-125 Seed with Sustained Self-Oxygenation for Sensitized Tumor Brachytherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411193. [PMID: 40026030 DOI: 10.1002/smll.202411193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Iodine-125 (125I) brachytherapy (BT) is renowned for its precision and effectiveness in delivering localized radiation doses to solid tumors. However, the therapeutic efficacy of traditional125I seed is often limited due to the inherent and acquired radioresistance. Based on the importance of tumor hypoxia in radioresistance, a novel "in situ oxygen-supplement" surface-modified radioactive 125I seed (125I@TNT-CaO2) is designed and constructed to overcome hypoxia-induced radioresistance in tumor BT. Titanium dioxide nanotubes (TNTs) are modified on the titanium shell of traditional 125I seed and loaded with nanoscale calcium peroxide (CaO2), further leading to a sustained release of O2. This in situ oxygen delivery system sensitizes hypoxic tumor regions to 125I BT, significantly improving therapeutic efficacy by inducing more ROS generation and DNA damage. Both in vitro and in vivo experiments demonstrate enhanced tumor suppression and apoptosis, with elevated O2 levels further inhibiting hypoxia-inducible factor 1-alpha (HIF-1α) and its associated signaling pathways. This innovative 125I@TNT-CaO2 seed presents a promising paradigm to enhance the effectiveness of BT by reversing hypoxia-mediated resistance.
Collapse
Affiliation(s)
- Zhongkai Wang
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Cheng Feng
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Shuting Lu
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210009, P. R. China
| | - Yong Wang
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Ruiyang Suo
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
| | - Kaizhi Jia
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Tong Sun
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Junhao Mei
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Tian Huang
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Xiaxing Bai
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Qi Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210009, P. R. China
| | - Xijuan Yao
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Bo Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
| | - Jinhe Guo
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Jian Lu
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| |
Collapse
|
6
|
Zuo CJ, Tian J. Global trends and emerging research in nanotechnology for esophageal cancer: a comprehensive bibliometric analysis. Discov Oncol 2025; 16:262. [PMID: 40029466 DOI: 10.1007/s12672-025-02018-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Despite the growing body of research on nanotechnology for esophageal cancer (EC), a comprehensive bibliometric analysis in this field has yet to be conducted. This study aims to fill this gap by analyzing global research trends, key contributors, and emerging themes in nanotechnology for EC. METHODS A bibliometric analysis was performed on publications from 1980 to 2024, using data from the Web of Science Core Collection. The analysis was conducted using VOSviewer, CiteSpace, and the R package 'bibliometrix' to visualize research trends, collaboration networks, and thematic areas. RESULTS The analysis included 419 documents authored by 2952 researchers from 44 countries. A significant increase in publications was observed, particularly after 2011, with China, the United States, and Japan leading the contributions. Prominent institutions, including Zhengzhou University and the Chinese Academy of Sciences, were identified as key players. The research predominantly focused on drug delivery systems, nanomedicine, and cancer treatment mechanisms, with emerging trends in the development of advanced nanomaterials for personalized therapies. CONCLUSION This comprehensive bibliometric analysis of nanotechnology applications in EC highlights global research trends, key contributors, and emerging research areas. The findings underscore the crucial role of nanotechnology in advancing treatment strategies for EC and identify areas for future research and interdisciplinary collaboration.
Collapse
Affiliation(s)
- Chun-Jian Zuo
- Department of Thoracic Surgery, Army Medical Center of People'S Liberation Army of China (PLA), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China.
| | - Jie Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Chengdu, 610041, China.
- Lung Cancer Center, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Chengdu, 610041, China.
| |
Collapse
|
7
|
Sun Y, Cao Z, Zhang X, Zhu X, Xu Z, Zhou H, Wei X, Du W, Xu L. Rod-Shaped Au@Ce Nano-Platforms for Enhancing Photodynamic Tumor Collaborative Therapy. SMALL METHODS 2025; 9:e2400945. [PMID: 39097952 DOI: 10.1002/smtd.202400945] [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: 06/25/2024] [Revised: 07/22/2024] [Indexed: 08/06/2024]
Abstract
Tumor photodynamic therapy (PDT) relies on intratumoral free radicals, while the limited oxygen source and the depletion of tissue oxygen may exacerbate the hypoxia. As the treatment progresses, there will eventually be a problem of insufficient free radicals. Here, it is found that Au@CeO2 nano-rods (Au@Ce NRs), assembled by gold nano-rods (Au NRs) and ceria nanoparticles (CeO2 NPs), can efficaciously absorb near-infrared light (NIR) to promote the release of oxygen and free radicals. Au@Ce NRs exhibit a higher proportion of Ce3+ (Ce2O3) after oxygen release, while Ce3+ is subsequently oxidized to Ce4+ (CeO2) by trace H2O2. Interestingly, Au@Ce NRs re-oxidized by trace H2O2 can re-release oxygen and free radicals again upon NIR treatment, achieving oxygenation/oxygen evolution, similar to charging/discharging. This loop maximizes the conversion of limited oxygen source into highly cytotoxic free radicals. As a result, when B16-F10 cells are treated by NIR/Au@Ce NRs, more tumor cells undergo apoptosis, consistent with the higher level of free radicals. Importantly, NIR/Au@Ce NRs successfully suppresses tumor growth and promotes the generation of epidermal collagen fibers in the transplanted tumor model. Therefore, the rod-shaped Au@Ce NRs provide an ideal platform for maximizing the utilization of intratumoral oxygen sources and improving the treatment of melanoma.
Collapse
Affiliation(s)
- Yuxiang Sun
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
- Department of Periodontology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, 210008, P. R. China
| | - Ziqi Cao
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital, School of Medicine, Shanghai Jiaotong University, No.600, Yishan Road, Xuhui, Shanghai, 200233, P. R. China
| | - Xiaoli Zhang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Xingchen Zhu
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Zhenyang Xu
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Hantong Zhou
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Xiaoer Wei
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital, School of Medicine, Shanghai Jiaotong University, No.600, Yishan Road, Xuhui, Shanghai, 200233, P. R. China
| | - Wenxian Du
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital, School of Medicine, Shanghai Jiaotong University, No.600, Yishan Road, Xuhui, Shanghai, 200233, P. R. China
| | - Li Xu
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| |
Collapse
|
8
|
Liu P, Hao L, Hsu JC, Zhou M, Luo Z, Peng Y, Cai W, Hu S. Biomineralized Nanocomposite-Integrated Microneedle Patch for Combined Brachytherapy and Photothermal Therapy in Postoperative Melanoma Recurrence and Infectious Wound Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414468. [PMID: 39903769 PMCID: PMC11948049 DOI: 10.1002/advs.202414468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/16/2025] [Indexed: 02/06/2025]
Abstract
In the surgical management of malignant melanoma, incomplete tumor resection and large-area cutaneous defects are major contributors to high locoregional recurrence and uncontrolled wound infections, resulting in poor prognosis and prolonged recovery times for patients. Herein, a versatile nanocomposite microneedle patch (referred to as GM-Ag2S/Ca32P) is designed to simultaneously eliminate residual tumor post-surgery and promote the healing of infectious wounds. This microneedle patch effectively penetrates subcutaneous tissues, delivering therapeutic payloads to infiltrating tumor cells and bacteria. The Ag2S/Ca32P nanocomposites encapsulated within the microneedle patch decompose in the acidic microenvironment of tumors and bacterial biofilms, releasing radioactive 32P and Ag2S nanodots, which enhance tumor eradication and bacteria killing through synergistic brachytherapy and photothermal therapy (PTT). Moreover, the nanocomposite microneedle patch promotes scar-free wound healing by reducing inflammation, and promoting granulation tissue formation, collagen deposition, and angiogenesis, thanks to localized hyperthermia, radiation, and the swelling and biodegradation of the microneedle matrices. This microneedle patch-based postoperative adjuvant therapy offers a comprehensive strategy for addressing melanoma recurrence and infectious wound healing, with promising potential for clinical application in postsurgical management.
Collapse
Affiliation(s)
- Peng Liu
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityNo. 87 Xiangya RoadChangshaHunan410008China
- Key Laboratory of Biological NanotechnologyNHC. No. 87 Xiangya RoadChangshaHunan410008China
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin‐MadisonMadisonWI53705USA
| | - Lu Hao
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityNo. 87 Xiangya RoadChangshaHunan410008China
| | - Jessica C. Hsu
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin‐MadisonMadisonWI53705USA
| | - Ming Zhou
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityNo. 87 Xiangya RoadChangshaHunan410008China
| | - Zhisheng Luo
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityNo. 87 Xiangya RoadChangshaHunan410008China
| | - Ying Peng
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Weibo Cai
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin‐MadisonMadisonWI53705USA
| | - Shuo Hu
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityNo. 87 Xiangya RoadChangshaHunan410008China
- Key Laboratory of Biological NanotechnologyNHC. No. 87 Xiangya RoadChangshaHunan410008China
| |
Collapse
|
9
|
Zhang Q, Ma RF, Ren TJ, Ren XY, Xu ZR. Engineering small extracellular vesicles with multivalent DNA probes for precise tumor targeting and enhanced synergistic therapy. J Colloid Interface Sci 2025; 679:335-348. [PMID: 39366263 DOI: 10.1016/j.jcis.2024.09.224] [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: 07/23/2024] [Revised: 08/31/2024] [Accepted: 09/27/2024] [Indexed: 10/06/2024]
Abstract
Small extracellular vesicles (sEVs) have gained wide attention as efficient carriers for disease treatment. However, the proclivity of sEVs to be ingested by source cells is insufficient to accurately target specific sites, posing a challenge in realizing controlled targeting treatment. Here, we developed an engineered sEV nanocarrier capable of precise tumor targeting and enhanced synergistic therapy. Multivalent DNA probes, comprising abundant AS1411 aptamers and telomerase primers, were innovatively modified on the sEV membrane (M-D-sEV) for precise tumor targeting. To achieve synergistic therapy, gold nanorod-cerium oxide nanostructures (Au NRs-CeO2) and manganese dioxide nanosheets-doxorubicin (MnO2 NSs-DOX) were encapsulated into liposomes (Lip-Mat). Then M-D-sEV and Lip-Mat were fused together through membrane fusion to obtain nanocarriers. Owing to the multivalence of the probes, the surface of the nanocarriers was loaded with numerous aptamers, which greatly enhances their targeting ability and promotes the accumulation of drugs. When nanocarriers were ingested by tumor cells, telomerase and multivalent DNA probes triggered their aggregation, enhancing the therapeutic effect. Furthermore, under laser irradiation, Au NRs-CeO2 converted light into hyperthermia, thereby inducing the destruction of nanocarriers membrane. This process initiated a series of reactions involving glutathione and H2O2 consumption, as well as DOX release, ultimately achieving synergistic tumor therapy. In vitro and in vivo studies demonstrated the remarkable targeting ability of multivalent DNA probes and excellent therapeutic effect of this strategy. The engineered strategy of sEVs provide a promising approach for precise tumor therapy and hold great potential for the development of efficient, safe, and personalized drug delivery systems.
Collapse
Affiliation(s)
- Qi Zhang
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Ruo-Fei Ma
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Ting-Ju Ren
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Xiu-Yan Ren
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Zhang-Run Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China.
| |
Collapse
|
10
|
Lan Y, Zhao S, Hou T, Ren Y, Tang J, Yin S, Wu Y. Mechanism of HIF-1α promoting proliferation, invasion and metastasis of nasopharyngeal carcinoma by regulating MMP-2 in hypoxic microenvironment. Heliyon 2024; 10:e40760. [PMID: 39691198 PMCID: PMC11650291 DOI: 10.1016/j.heliyon.2024.e40760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Objective To explore the mechanism of HIF-1α promoting the proliferation, invasion and metastasis of nasopharyngeal carcinoma cells by regulating the expression of MMP-2. Methods 30 nasopharyngeal carcinoma tissues and 30 normal nasopharyngeal epithelial tissues were collected, and the expression of HIF-1α and MMP-2 in the nasopharyngeal carcinoma, normal nasopharyngeal epithelial tissues and their hypoxic environment were systematically analyzed by qRT-PCR and western blot techniques. Lentivirus transfection technology was used to regulate the expression of HIF-1α and MMP-2 genes in the HONE1 cell line under hypoxic environment, and to explore the interaction mechanism of HIF-1α and MMP-2 genes and their role in the proliferation, invasion and metastasis of nasopharyngeal carcinoma. Furthermore, the cytological behavior changes regulated by HIF-1α and MMP-2 genes were further explored by gene chip technology. Results The expressions of HIF-1α and MMP-2 in nasopharyngeal carcinoma tissues were significantly higher than those in normal nasopharyngeal epithelial tissues (P < 0.05). Compared with normoxic group, the expression of HIF-1α and MMP-2 in the nasopharyngeal carcinoma cell line HONE1 increased in hypoxic group (P < 0.05). Compared with NC-siRNA group, the expression of HIF-1α in si-HIF-1α group decreased, and the cell proliferation ability and invasion and metastasis ability decreased (P < 0.05). PCR array analysis revealed that the mRNA expressions of FAS, BRCA1, TIMP-1 genes were up-regulated in nasopharyngeal carcinoma HONE1 cells with HIF-1α gene silencing. AKT1, VEGFA, MET, MMP-2, MMP-9 and MTA2 were down-regulated. Compared with NC-siRNA group, the expression of MMP-2 in si-MMP-2 group decreased, and the ability of cell proliferation and invasion and metastasis decreased (P < 0.05). Conclusion HIF-1α could inhibit the proliferation, invasion and metastasis of nasopharyngeal carcinoma by regulating the expression of MMP-2, thus inhibiting tumor growth. Therefore, HIF-1α and MMP-2 might become important therapeutic targets to inhibit the growth, invasion and metastasis of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Ying Lan
- Department of Otolaryngology Head and Neck Surgery, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shijie Zhao
- Department of Otolaryngology Head and Neck Surgery, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Tao Hou
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yi Ren
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jun Tang
- Department of Otolaryngology Head and Neck Surgery, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shihua Yin
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yang Wu
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
11
|
Mi L, Zhang H. Myriad factors and pathways influencing tumor radiotherapy resistance. Open Life Sci 2024; 19:20220992. [PMID: 39655194 PMCID: PMC11627069 DOI: 10.1515/biol-2022-0992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/09/2024] [Accepted: 09/30/2024] [Indexed: 12/12/2024] Open
Abstract
Radiotherapy is a cornerstone in the treatment of various tumors, yet radioresistance often leads to treatment failure and tumor recurrence. Several factors contribute to this resistance, including hypoxia, DNA repair mechanisms, and cancer stem cells. This review explores the diverse elements that drive tumor radiotherapy resistance. Historically, resistance has been attributed to cellular repair and tumor repopulation, but recent research has expanded this understanding. The tumor microenvironment - characterized by hypoxia, immune evasion, and stromal interactions - further complicates treatment. Additionally, molecular mechanisms such as aberrant signaling pathways, epigenetic modifications, and non-B-DNA structures play significant roles in mediating resistance. This review synthesizes current knowledge, highlighting the interplay of these factors and their clinical implications. Understanding these mechanisms is crucial for developing strategies to overcome resistance and improve therapeutic outcomes in cancer patients.
Collapse
Affiliation(s)
- Lanjuan Mi
- School of Life and Health Sciences, Huzhou College, Hu Zhou, China
| | - Hongquan Zhang
- The First Affiliated Hospital of Huzhou University, Hu Zhou, China
| |
Collapse
|
12
|
Wang L, Zhang L, Zhang Z, Wu P, Zhang Y, Chen X. Advances in targeting tumor microenvironment for immunotherapy. Front Immunol 2024; 15:1472772. [PMID: 39421736 PMCID: PMC11484021 DOI: 10.3389/fimmu.2024.1472772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The tumor microenvironment (TME) provides essential conditions for the occurrence, invasion, and spread of cancer cells. Initial research has uncovered immunosuppressive properties of the TME, which include low oxygen levels (hypoxia), acidic conditions (low pH), increased interstitial pressure, heightened permeability of tumor vasculature, and an inflammatory microenvironment. The presence of various immunosuppressive components leads to immune evasion and affects immunotherapy efficacy. This indicates the potential value of targeting the TME in cancer immunotherapy. Therefore, TME remodeling has become an effective method for enhancing host immune responses against tumors. In this study, we elaborate on the characteristics and composition of the TME and how it weakens immune surveillance and summarize targeted therapeutic strategies for regulating the TME.
Collapse
Affiliation(s)
- Lugang Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liubo Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Wu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
13
|
Zhang TQ, Lv QY, Jin WL. The cellular-centered view of hypoxia tumor microenvironment: Molecular mechanisms and therapeutic interventions. Biochim Biophys Acta Rev Cancer 2024; 1879:189137. [PMID: 38880161 DOI: 10.1016/j.bbcan.2024.189137] [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: 12/11/2023] [Revised: 06/01/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Cancer is a profoundly dynamic, heterogeneous and aggressive systemic ailment, with a coordinated evolution of various types of tumor niches. Hypoxia plays an indispensable role in the tumor micro-ecosystem, drastically enhancing the plasticity of cancer cells, fibroblasts and immune cells and orchestrating intercellular communication. Hypoxia-induced signals, particularly hypoxia-inducible factor-1α (HIF-1α), drive the reprogramming of genetic, transcriptional, and proteomic profiles. This leads to a spectrum of interconnected processes, including augmented survival of cancer cells, evasion of immune surveillance, metabolic reprogramming, remodeling of the extracellular matrix, and the development of resistance to conventional therapeutic modalities like radiotherapy and chemotherapy. Here, we summarize the latest research on the multifaceted effects of hypoxia, where a multitude of cellular and non-cellular elements crosstalk with each other and co-evolve in a synergistic manner. Additionally, we investigate therapeutic approaches targeting hypoxic niche, encompassing hypoxia-activated prodrugs, HIF inhibitors, nanomedicines, and combination therapies. Finally, we discuss some of the issues to be addressed and highlight the potential of emerging technologies in the treatment of cancer.
Collapse
Affiliation(s)
- Tian-Qi Zhang
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; The Second Hospital of Jilin University, Changchun 130041, China
| | - Qian-Yu Lv
- The Second Hospital of Jilin University, Changchun 130041, China
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
14
|
Li Q, Lin G, Zhang K, Liu X, Li Z, Bing X, Nie Z, Jin S, Guo J, Min X. Hypoxia exposure induces lactylation of Axin1 protein to promote glycolysis of esophageal carcinoma cells. Biochem Pharmacol 2024; 226:116415. [PMID: 38972426 DOI: 10.1016/j.bcp.2024.116415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
The hypoxic microenvironment in esophageal carcinoma is an important factor promoting the rapid progression of malignant tumor. This study was to investigate the lactylation of Axin1 on glycolysis in esophageal carcinoma cells under hypoxia exposure. Hypoxia treatment increases pan lysine lactylation (pan-kla) levels of both TE1 and EC109 cells. Meanwhile, ECAR, glucose consumption and lactate production were also upregulated in both TE1 and EC109 cells. The expression of embryonic stem cell transcription factors NANOG and SOX2 were enhanced in the hypoxia-treated cells. Axin1 overexpression partly reverses the induction effects of hypoxia treatment in TE1 and EC109 cells. Moreover, lactylation of Axin1 protein at K147 induced by hypoxia treatment promotes ubiquitination modification of Axin1 protein to promote glycolysis and cell stemness of TE1 and EC109 cells. Mutant Axin1 can inhibit ECAR, glucose uptake, lactate secretion, and cell stemness in TE1 and EC109 cells under normal or hypoxia conditions. Meanwhile, mutant Axin1 further enhanced the effects of 2-DG on inhibiting glycolysis and cell stemness. Overexpression of Axin1 also inhibited tumor growth in vivo, and was related to suppressing glycolysis. In conclusion, hypoxia treatment promoted the glycolysis and cell stemness of esophageal carcinoma cells, and increased the lactylation of Axin1 protein. Overexpression of Axin1 functioned as a glycolysis inhibitor, and suppressed the effects of hypoxia exposure in vitro and inhibited tumor growth in vivo. Mechanically, hypoxia induces the lactylation of Axin1 protein and promotes the ubiquitination of Axin1 to degrade the protein, thereby exercising its anti-glycolytic function.
Collapse
Affiliation(s)
- Qian Li
- Department of Thoracic Surgery, China Aerospace Science and Industry Corporation 731 Hospital, Beijing, China; Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guihu Lin
- Department of Thoracic Surgery, China Aerospace Science and Industry Corporation 731 Hospital, Beijing, China
| | - Kaihua Zhang
- Department of Thoracic Surgery, China Aerospace Science and Industry Corporation 731 Hospital, Beijing, China
| | - Xinbo Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhantao Li
- Department of Thoracic Surgery, China Aerospace Science and Industry Corporation 731 Hospital, Beijing, China
| | - Xiaohan Bing
- Department of Thoracic Surgery, China Aerospace Science and Industry Corporation 731 Hospital, Beijing, China
| | - Zhenkai Nie
- Department of Thoracic Surgery, China Aerospace Science and Industry Corporation 731 Hospital, Beijing, China
| | - Shan Jin
- Department of Thoracic Surgery, China Aerospace Science and Industry Corporation 731 Hospital, Beijing, China
| | - Jin Guo
- School of Medical Technology, Beijing Institute of Technology, Beijing, China.
| | - Xianjun Min
- Department of Thoracic Surgery, China Aerospace Science and Industry Corporation 731 Hospital, Beijing, China.
| |
Collapse
|
15
|
Lu X, Zhu Y, Deng X, Kong F, Xi C, Luo Q, Zhu X. Development of a Supermolecular Radionuclide-Drug Conjugate System for Integrated Radiotheranostics for Non-small Cell Lung Cancer. J Med Chem 2024; 67:11152-11167. [PMID: 38896797 DOI: 10.1021/acs.jmedchem.4c00673] [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: 06/21/2024]
Abstract
Radionuclide-drug conjugates (RDCs) designed from small molecule or nanoplatform shows complementary characteristics. We constructed a new RDC system with integrated merits of small molecule and nanoplatform-based RDCs. Erlotinib was labeled with 131I to construct the bulk of RDC (131I-ER). Floxuridine was mixed with 131I-ER to develop a hydrogen bond-driving supermolecular RDC system (131I-ER-Fu NPs). The carrier-free 131I-ER-Fu NPs supermolecule not only demonstrated integrated merits of small molecule and nanoplatform-based RDC, including clear structure definition, stable quality control, prolonged circulation lifetime, enhanced tumor specificity and retention, and rapidly nontarget clearance, but also exhibited low biological toxicity and stronger antitumor effects. In vivo imaging also revealed its application for tumor localization of nonsmall cell lung cancer (NSCLC) and screening of patients suitable for epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) therapy. We considered that 131I-ER-Fu NPs showed potentials as an integrated platform for the radiotheranostics of NSCLC.
Collapse
Affiliation(s)
- Xinmiao Lu
- Department of Nuclear Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Yunyun Zhu
- Department of Nuclear Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Xiaohui Deng
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fei Kong
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chuang Xi
- Department of Nuclear Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Quanyong Luo
- Department of Nuclear Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
16
|
Xie H, Xiao X, Yi X, Huang K, Wang L. Cell-Membrane-Coated Metal-Organic Framework Nanocarrier Combining Chemodynamic Therapy for the Inhibition of Hepatocellular Carcinoma Proliferation. Pharmaceutics 2024; 16:619. [PMID: 38794281 PMCID: PMC11124917 DOI: 10.3390/pharmaceutics16050619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Chemodynamic therapy (CDT) employs hydrogen peroxide (H2O2) within the tumor microenvironment (TME) to initiate the Fenton reaction and catalyze the generation of hydroxyl radicals (·OH) for targeted therapy. Metal ion-based nanomaterials have garnered significant attention as catalysts due to their potent anti-tumor effects. Hypoxia in the TME is often associated with cancer cell development and metastasis, with HIF-1α being a pivotal factor in hypoxia adaptation. In this study, an organic framework called MIL-101 (Fe) was designed and synthesized to facilitate H2O2-induced ·OH production while also serving as a carrier for the HIF-1α inhibitor Acriflavine (ACF). A biomimetic nanomedical drug delivery system named MIL-101/ACF@CCM was constructed by encapsulating liver cancer cell membranes onto the framework. This delivery system utilized the homologous targeting of tumor cell membranes to transport ACF, inhibiting HIF-1α expression, alleviating tumor hypoxia, and catalyzing ·OH production for effective tumor eradication. Both in vivo and in vitro experiments confirmed that combining ACF with chemotherapy achieved remarkable tumor inhibition by enhancing ROS production and suppressing HIF-1α expression.
Collapse
Affiliation(s)
- Huaying Xie
- The First School of Clinical Medicine, Guilin Medical University, Guilin 541006, China; (H.X.); (X.Y.); (K.H.)
| | - Xuhua Xiao
- Department of Gastroenterology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China;
| | - Xiaoyuan Yi
- The First School of Clinical Medicine, Guilin Medical University, Guilin 541006, China; (H.X.); (X.Y.); (K.H.)
| | - Kunzhao Huang
- The First School of Clinical Medicine, Guilin Medical University, Guilin 541006, China; (H.X.); (X.Y.); (K.H.)
| | - Liyan Wang
- Department of Gastroenterology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China;
| |
Collapse
|
17
|
Li Z, Liu X, Xiao J, Jiang H, Ma L, Luo Y, Wang M, Zhu Y, Jiang H, Yao H, Ngai T, Guo Q. Ultrastable Iodinated Oil-Based Pickering Emulsion Enables Locoregional Sustained Codelivery of Hypoxia Inducible Factor-1 Inhibitor and Anticancer Drugs for Tumor Combination Chemotherapy. ACS Biomater Sci Eng 2024; 10:2270-2281. [PMID: 38536862 DOI: 10.1021/acsbiomaterials.3c01887] [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] [Indexed: 04/09/2024]
Abstract
Tumor hypoxia-associated drug resistance presents a major challenge for cancer chemotherapy. However, sustained delivery systems with a high loading capability of hypoxia-inducible factor-1 (HIF-1) inhibitors are still limited. Here, we developed an ultrastable iodinated oil-based Pickering emulsion (PE) to achieve locally sustained codelivery of a HIF-1 inhibitor of acriflavine and an anticancer drug of doxorubicin for tumor synergistic chemotherapy. The PE exhibited facile injectability for intratumoral administration, great radiopacity for in vivo examination, excellent physical stability (>1 mo), and long-term sustained release capability of both hydrophilic drugs (i.e., acriflavine and doxorubicin). We found that the codelivery of acriflavine and doxorubicin from the PE promoted the local accumulation and retention of both drugs using an acellular liver organ model and demonstrated significant inhibition of tumor growth in a 4T1 tumor-bearing mouse model, improving the chemotherapeutic efficacy through the synergistic effects of direct cytotoxicity with the functional suppression of HIF-1 pathways of tumor cells. Such an iodinated oil-based PE provides a great injectable sustained delivery platform of hydrophilic drugs for locoregional chemotherapy.
Collapse
Affiliation(s)
- Zhihua Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaoya Liu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jingyu Xiao
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hang Jiang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Le Ma
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yucheng Luo
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Meijuan Wang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuwei Zhu
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, PR China
| | - Hongliang Jiang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hanyang Yao
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - To Ngai
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, PR China
| | - Qiongyu Guo
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
18
|
Li A, Yang J, He Y, Wen J, Jiang X. Advancing piezoelectric 2D nanomaterials for applications in drug delivery systems and therapeutic approaches. NANOSCALE HORIZONS 2024; 9:365-383. [PMID: 38230559 DOI: 10.1039/d3nh00578j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Precision drug delivery and multimodal synergistic therapy are crucial in treating diverse ailments, such as cancer, tissue damage, and degenerative diseases. Electrodes that emit electric pulses have proven effective in enhancing molecule release and permeability in drug delivery systems. Moreover, the physiological electrical microenvironment plays a vital role in regulating biological functions and triggering action potentials in neural and muscular tissues. Due to their unique noncentrosymmetric structures, many 2D materials exhibit outstanding piezoelectric performance, generating positive and negative charges under mechanical forces. This ability facilitates precise drug targeting and ensures high stimulus responsiveness, thereby controlling cellular destinies. Additionally, the abundant active sites within piezoelectric 2D materials facilitate efficient catalysis through piezochemical coupling, offering multimodal synergistic therapeutic strategies. However, the full potential of piezoelectric 2D nanomaterials in drug delivery system design remains underexplored due to research gaps. In this context, the current applications of piezoelectric 2D materials in disease management are summarized in this review, and the development of drug delivery systems influenced by these materials is forecast.
Collapse
Affiliation(s)
- Anshuo Li
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, No. 639 Zhizaoju Road, Shanghai 200011, China.
- State Key Laboratory of Metastable Materials Science and Technology, Nanobiotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao, 066004, China
| | - Jiawei Yang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, No. 639 Zhizaoju Road, Shanghai 200011, China.
| | - Yuchu He
- State Key Laboratory of Metastable Materials Science and Technology, Nanobiotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao, 066004, China
| | - Jin Wen
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, No. 639 Zhizaoju Road, Shanghai 200011, China.
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, No. 639 Zhizaoju Road, Shanghai 200011, China.
| |
Collapse
|
19
|
Li J, Wang T, Shi Y, Ye Z, Zhang X, Ming J, Zhang Y, Hu X, Li Y, Zhang D, Xu Q, Yang J, Chen X, Liu N, Su X. A continuously efficient O 2-supplying strategy for long-term modulation of hypoxic tumor microenvironment to enhance long-acting radionuclides internal therapy. J Nanobiotechnology 2024; 22:7. [PMID: 38166931 PMCID: PMC10763042 DOI: 10.1186/s12951-023-02268-5] [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: 11/18/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Radionuclides internal radiotherapy (RIT) is a clinically powerful method for cancer treatment, but still poses unsatisfactory therapeutic outcomes due to the hypoxic characteristic of tumor microenvironment (TME). Catalase (CAT) or CAT-like nanomaterials can be used to enzymatically decompose TME endogenous H2O2 to boost TME oxygenation and thus alleviate the hypoxic level within tumors, but their effectiveness is still hindered by the short-lasting of hypoxia relief owing to their poor stability or degradability, thereby failing to match the long therapeutic duration of RIT. Herein, we proposed an innovative strategy of using facet-dependent CAT-like Pd-based two-dimensional (2D) nanoplatforms to continuously enhance RIT. Specifically, rationally designed 2D Pd@Au nanosheets (NSs) enable consistent enzymatic conversion of endogenous H2O2 into O2 to overcome hypoxia-induced RIT resistance. Furthermore, partially coated Au layer afford NIR-II responsiveness and moderate photothermal treatment that augmenting their enzymatic functionality. This approach with dual-effect paves the way for reshaping TME and consequently facilitating the brachytherapy ablation of cancer. Our work offers a significant advancement in the integration of catalytic nanomedicine and nuclear medicine, with the overarching goal of amplifying the clinical benefits of RIT-treated patients.
Collapse
Affiliation(s)
- Jingchao Li
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tingting Wang
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuanfei Shi
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zichen Ye
- State Key Laboratory for Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, and Engineering Research Center for Nano-Preparation Technology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Xun Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiang Ming
- State Key Laboratory for Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, and Engineering Research Center for Nano-Preparation Technology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Yafei Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xinyan Hu
- State Key Laboratory for Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, and Engineering Research Center for Nano-Preparation Technology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Yun Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361005, China
| | - Dongsheng Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qianhe Xu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jun Yang
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xiaolan Chen
- State Key Laboratory for Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, and Engineering Research Center for Nano-Preparation Technology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| | - Nian Liu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Xinhui Su
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
20
|
He S, Gou X, Zhang S, Zhang X, Huang H, Wang W, Yi L, Zhang R, Duan Z, Zhou P, Qian Z, Gao X. Nanodelivery Systems as a Novel Strategy to Overcome Treatment Failure of Cancer. SMALL METHODS 2024; 8:e2301127. [PMID: 37849248 DOI: 10.1002/smtd.202301127] [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: 08/24/2023] [Revised: 09/25/2023] [Indexed: 10/19/2023]
Abstract
Despite the tremendous progress in cancer treatment in recent decades, cancers often become resistant due to multiple mechanisms, such as intrinsic or acquired multidrug resistance, which leads to unsatisfactory treatment effects or accompanying metastasis and recurrence, ultimately to treatment failure. With a deeper understanding of the molecular mechanisms of tumors, researchers have realized that treatment designs targeting tumor resistance mechanisms would be a promising strategy to break the therapeutic deadlock. Nanodelivery systems have excellent physicochemical properties, including highly efficient tissue-specific delivery, substantial specific surface area, and controllable surface chemistry, which endow nanodelivery systems with capabilities such as precise targeting, deep penetration, responsive drug release, multidrug codelivery, and multimodal synergy, which are currently widely used in biomedical researches and bring a new dawn for overcoming cancer resistance. Based on the mechanisms of tumor therapeutic resistance, this review summarizes the research progress of nanodelivery systems for overcoming tumor resistance to improve therapeutic efficacy in recent years and offers prospects and challenges of the application of nanodelivery systems for overcoming cancer resistance.
Collapse
Affiliation(s)
- Shi He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xinyu Gou
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Shuheng Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xifeng Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Hongyi Huang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Wanyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Linbin Yi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Rui Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhongxin Duan
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Peizhi Zhou
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| |
Collapse
|
21
|
An L, Li M, Jia Q. Mechanisms of radiotherapy resistance and radiosensitization strategies for esophageal squamous cell carcinoma. Mol Cancer 2023; 22:140. [PMID: 37598158 PMCID: PMC10439611 DOI: 10.1186/s12943-023-01839-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/02/2023] [Indexed: 08/21/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the sixth most common cause of cancer-related mortality worldwide, with more than half of them occurred in China. Radiotherapy (RT) has been widely used for treating ESCC. However, radiation-induced DNA damage response (DDR) can promote the release of cytokines and chemokines, and triggers inflammatory reactions and changes in the tumor microenvironment (TME), thereby inhibiting the immune function and causing the invasion and metastasis of ESCC. Radioresistance is the major cause of disease progression and mortality in cancer, and it is associated with heterogeneity. Therefore, a better understanding of the radioresistance mechanisms may generate more reversal strategies to improve the cure rates and survival periods of ESCC patients. We mainly summarized the possible mechanisms of radioresistance in order to reveal new targets for ESCC therapy. Then we summarized and compared the current strategies to reverse radioresistance.
Collapse
Affiliation(s)
- Lingbo An
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- College of Medical Technology, Xi'an Medical University, Xi'an, China
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| |
Collapse
|
22
|
Zhang L, Li X, Yue G, Guo L, Hu Y, Cui Q, Wang J, Tang J, Liu H. Nanodrugs systems for therapy and diagnosis of esophageal cancer. Front Bioeng Biotechnol 2023; 11:1233476. [PMID: 37520291 PMCID: PMC10373894 DOI: 10.3389/fbioe.2023.1233476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 06/29/2023] [Indexed: 08/01/2023] Open
Abstract
With the increasing incidence of esophageal cancer, its diagnosis and treatment have become one of the key issues in medical research today. However, the current diagnostic and treatment methods face many unresolved issues, such as low accuracy of early diagnosis, painful treatment process for patients, and high recurrence rate after recovery. Therefore, new methods for the diagnosis and treatment of esophageal cancer need to be further explored, and the rapid development of nanomaterials has brought new ideas for solving this problem. Nanomaterials used as drugs or drug delivery systems possess several advantages, such as high drug capacity, adjustably specific targeting capability, and stable structure, which endow nanomaterials great application potential in cancer therapy. However, even though the nanomaterials have been widely used in cancer therapy, there are still few reviews on their application in esophageal cancer, and systematical overview and analysis are deficient. Herein, we overviewed the application of nanodrug systems in therapy and diagnosis of esophageal cancer and summarized some representative case of their application in diagnosis, chemotherapy, targeted drug, radiotherapy, immunity, surgery and new therapeutic method of esophageal cancer. In addition, the nanomaterials used for therapy of esophageal cancer complications, esophageal stenosis or obstruction and oesophagitis, are also listed here. Finally, the challenge and the future of nanomaterials used in cancer therapy were discussed.
Collapse
Affiliation(s)
- Lihan Zhang
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Xing Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Guangxing Yue
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Lihao Guo
- Interdisciplinary Research Center of Smart Sensors, School of Advanced Materials and Nanotechnology, Xidian University, Xi’an, China
| | - Yanhui Hu
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Qingli Cui
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jia Wang
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jingwen Tang
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Huaimin Liu
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
23
|
Li X, Wang H, Li Z, Tao F, Wu J, Guan W, Liu S. Oxygen switches: Refueling for cancer radiotherapy. Front Oncol 2023; 12:1085432. [PMID: 36873299 PMCID: PMC9978393 DOI: 10.3389/fonc.2022.1085432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/28/2022] [Indexed: 02/18/2023] Open
Abstract
Radiotherapy remains the major therapeutic intervention for tumor patients. However, the hypoxic tumor microenvironment leads to treatment resistance. Recently, a burgeoning number of nano-radiosensitizers designed to increase the oxygen concentration in tumors were reported. These nano radiosensitizers served as oxygen carriers, oxygen generators, and even sustained oxygen pumps, attracting increased research interest. In this review, we focus on the novel oxygen-enrich nano radiosensitizers, which we call oxygen switches, and highlight their influence in radiotherapy through different strategies. Physical strategies-based oxygen switches carried O2 into the tumor via their high oxygen capacity. The chemical reactions to generate O2 in situ were triggered by chemical strategies-based oxygen switches. Biological strategies-based oxygen switches regulated tumor metabolism, remodeled tumor vasculature, and even introduced microorganisms-mediated photosynthesis for long-lasting hypoxia alleviating. Moreover, the challenges and perspectives of oxygen switches-mediated oxygen-enrich radiotherapy were discussed.
Collapse
Affiliation(s)
- Xianghui Li
- First Affiliated Hospital of Guangxi Medical University, Depatment of Dermatology, Nanning, China
- Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing, China
| | - Haoran Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhiyan Li
- First Affiliated Hospital of Guangxi Medical University, Depatment of Dermatology, Nanning, China
| | - Feng Tao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing, China
| | - Wenxian Guan
- First Affiliated Hospital of Guangxi Medical University, Depatment of Dermatology, Nanning, China
| | - Song Liu
- First Affiliated Hospital of Guangxi Medical University, Depatment of Dermatology, Nanning, China
| |
Collapse
|