1
|
Zhang H, Wang Y, Jiang M, Wang K, Yan J, Li G, Zheng Z. Inherently anti-metastatic peptide hydrogels for sonodynamic-amplified ferroptosis in cancer therapy. Mater Today Bio 2025; 32:101688. [PMID: 40206142 PMCID: PMC11980000 DOI: 10.1016/j.mtbio.2025.101688] [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: 01/13/2025] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer metastasis remains a significant challenge in oncology, prompting the exploration of innovative biomaterials to enhance treatment efficacy. While many hydrogels only serve as passive carriers, this study presents two novel self-assembling peptides, CWEWTWY and NapFFSGP, which form supramolecular hydrogels with intrinsic anti-metastatic properties. We demonstrate a correlation between the nanofibrous morphology of these peptides and their enhanced anti-metastatic activity, mediated by disruption of F-actin organization and impacting pathways related to cancer cell adhesion and actin filament dynamics. In vivo studies confirm a significant reduction in lung metastasis using a 4T1 pulmonary metastasis model. We also demonstrate their potential as a simple, synergistic platform integrating sonodynamic therapy (SDT) and ferroptosis. Ironporphyrin (FP), incorporated into Gel@FP, acts as both a sonosensitizer and ferroptosis inducer. Upon ultrasound irradiation, FP generates localized reactive oxygen species, further amplifying ferroptosis through enhanced lipid peroxidation. Gel@FP combined with ultrasound demonstrates potent antitumor efficacy in vitro and in vivo, promoting apoptosis, ferroptosis, and immunogenic cell death, leading to enhanced tumor regression and robust immune activation. Our findings highlight the potential of anti-metastatic hydrogels as a promising multifunctional platform to address the challenges of metastasis while enhancing antitumor immunity.
Collapse
Affiliation(s)
- Hongxia Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Yamei Wang
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Mengmeng Jiang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Kunyu Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Jingru Yan
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Gongyu Li
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhen Zheng
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
2
|
Zhu K, Wang J, Wang Z, Chen Q, Song J, Chen X. Ultrasound-Activated Theranostic Materials and Their Bioapplications. Angew Chem Int Ed Engl 2025; 64:e202422278. [PMID: 40091509 DOI: 10.1002/anie.202422278] [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: 11/15/2024] [Revised: 03/01/2025] [Accepted: 03/16/2025] [Indexed: 03/19/2025]
Abstract
Ultrasound (US) is a promising external excitation modality for bioapplications, offering significant advantages over X-rays or lasers due to its low cost, high biosafety, and ideal tissue penetration depth. US-activated theranostic materials, comprising organic, inorganic, and hybrid-based compounds, hold particular value in synergistic cancer therapeutic and diagnostic applications. These materials exhibit excellent imaging properties, high drug delivery and release efficiency, and enhanced reactive oxygen species (ROS) production, making them suitable for clinical diagnostic imaging and therapeutic interventions. This review summarizes recent research on the design, performance, and optimization of US-mediated molecules/nanosystems for a wide range of biomedical applications. Additionally, the multifunctional use of these sonosensitizers in imaging, drug delivery, and sonodynamic therapy, especially in combination with other treatments, could pave the way for innovative strategies in disease therapy. Finally, an overview of this field's challenges and potential future directions is provided, highlighting pathways to promote clinical translation and application.
Collapse
Affiliation(s)
- Kang Zhu
- State Key Laboratory of Chemical Resource Engineering, Cellege of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
| | - Jimei Wang
- State Key Laboratory of Chemical Resource Engineering, Cellege of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
| | - Zhao Wang
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Jinan, 250000, P.R. China
| | - Qing Chen
- State Key Laboratory of Chemical Resource Engineering, Cellege of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, Cellege of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
| | - Xiaoyuan Chen
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Lower Kent Ridge Road, 4 Science Drive 2, Singapore, 117544, Singapore
| |
Collapse
|
3
|
Xie M, Jiang C, Zhang C, Wu Y, Zhang X, Yao R, Han C, Dai Y, Xu K, Zheng S. Tumor microenvironment triggered iron-based metal organic frameworks for magnetic resonance imaging and photodynamic-enhanced ferroptosis therapy. J Colloid Interface Sci 2025; 685:382-395. [PMID: 39855085 DOI: 10.1016/j.jcis.2025.01.119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Photodynamic therapy (PDT) primarily relies on the generation of reactive oxygen species (ROS) to eliminate tumor cells. However, the elevated levels of glutathione (GSH) within tumor cells can limit the efficacy of PDT, posing a challenge to achieve complete tumor eradication. Herein, a porous iron-based metal-organic frameworks (PEG-Fe-MOFs) nanoplatform was developed for the combined application of PDT and ferroptosis in cancer treatment. The coordination between tetrakis (4-carboxyphenyl) porphyrin (TCPP) and ferric (Fe3+) enabled PEG-modified Fe-MOFs (PEG-Fe-MOFs) to deliver excellent T1-weighted magnetic resonance (MR) imaging performance in physiological environments. Within the tumor microenvironment (TME), PEG-Fe-MOFs gradually degraded to release TCPP, which could be utilized for fluorescence imaging. Moreover, Fe2+ enhanced intracellular ROS levels via the Fenton reaction, generating hydroxyl radicals that further amplified ROS production. This synergistic effect comprising increased ROS levels and GSH depletion augmented the efficacy of PDT while simultaneously inducing robust ferroptosis in tumor cells, thereby maximizing therapeutic outcomes. Both in vitro and in vivo experiments have demonstrated the superior T1 weighted MR and fluorescence imaging capabilities of PEG-Fe-MOFs, along with its potent synergistic therapeutic effects on tumors. These results highlighted the potential of this nanoplatform for combining PDT and ferroptosis in cancer treatment.
Collapse
Affiliation(s)
- Manman Xie
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Canran Jiang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Cong Zhang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Yun Wu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiuli Zhang
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Ruosi Yao
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Cuiping Han
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China.
| | - Yue Dai
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China.
| | - Kai Xu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China; Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China.
| | - Shaohui Zheng
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China; Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
4
|
Yu J, Hu JR, Tian Y, Lei YM, Hu HM, Lei BS, Zhang G, Sun Y, Ye HR. Nanosensitizer-assisted sonodynamic therapy for breast cancer. J Nanobiotechnology 2025; 23:281. [PMID: 40197318 PMCID: PMC11978163 DOI: 10.1186/s12951-025-03311-3] [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: 01/05/2025] [Accepted: 03/09/2025] [Indexed: 04/10/2025] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide. Despite advancements in therapeutic modalities, its prognosis remains poor owing to complex clinical, pathological, and molecular characteristics. Sonodynamic therapy (SDT) is a promising approach for tumor elimination, using sonosensitizers that preferentially accumulate in tumor tissues and are activated by low-intensity ultrasound to produce reactive oxygen species. However, the clinical translation of SDT faces challenges, including the limited efficiency of sonosensitizers and resistance posed by the tumor microenvironment. The emergence of nanomedicine offers innovative strategies to address these obstacles. This review discusses strategies for enhancing the efficacy of SDT using sonosensitizers, including rational structural modifications, improved tumor-targeted enrichment, tumor microenvironment remodeling, and imaging-guided therapy. Additionally, SDT-based multimodal therapies, such as sono-chemotherapy, sono-immunotherapy, and sono-photodynamic therapy, and their potential applications in breast cancer treatment are summarized. The underlying mechanisms of SDT in breast cancer are briefly outlined. Finally, this review highlights current challenges and prospects for the clinical translation of SDT, providing insights into future advancements that may improve therapeutic outcomes for breast cancer.
Collapse
Affiliation(s)
- Jing Yu
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Jun-Rui Hu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yi Tian
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Yu-Meng Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Hai-Man Hu
- Department of Electrical and Electronic Engineering, Hubei University of Technology, Wuhan, 430068, China
| | - Bing-Song Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China.
| | - Ge Zhang
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China.
| | - Yao Sun
- National Key Laboratory of Green Pesticides, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Hua-Rong Ye
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China.
| |
Collapse
|
5
|
Wang MF, Guo J, Yuan SJ, Li K, Zhang Q, Lei HM, Wu JL, Zhao L, Xu YH, Chen X. Targeted sonodynamic therapy induces tumor cell quasi-immunogenic ferroptosis and macrophage immunostimulatory autophagy in glioblastoma. Biomaterials 2025; 315:122913. [PMID: 39471712 DOI: 10.1016/j.biomaterials.2024.122913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/13/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
In this study, we demonstrated the mechanism of a glioblastoma (GBM)-targeted sonodynamic therapy (SDT) strategy employing platelets loaded with a sonosensitizer based on functionalized boron nitride nanoparticles carrying chlorin e6 (BNPD-Ce6). In the in vitro study, we first found that the BNPD-Ce6-mediated sonodynamic action (SDA) induced remarkable viability loss, DNA damage, and cell death in the GBM cells (GBCs) but not macrophages. Surprisingly, the SDA-exposed GBCs displayed a ferroptotic phenotype while the SDA-exposed macrophages underwent immuno-stimulatory autophagy and potently potentiated the SDA's toxicity to the GBCs. The ferroptotic GBCs induced by the SDA were found to be quasi-immunogenic, characterized by the emission of some alarmins such as ATP, HSP90, and CRT, but absent HMGB1, a potent endogenous adjuvant. As such, the SDA-stressed GBCs were unable to stimulate the BMDMs. This defect, interestingly, could be rescued by platelets as a donor of HMGB1 which markedly enhanced the BNPD-Ce6's sonotoxicity to the GBCs. In the in vivo study, we first employed BNPD-Ce6-loaded platelets to achieve ultrasound-triggered, targeted delivery of BNPD-Ce6 in grafted intra-cranial GBMs and subsequent sonodynamic tumor damage. An SDT regimen designed based on these results slowed the growth of grafted intra-cranial GBMs and significantly increased the survival of the host animals. Pathological examination of the SDT-treated GBMs revealed tissue necrosis and destruction and validated the in vitro observations. Finally, the depletion of macrophages was found to abrogate the efficacy of the SDT in subcutaneous GBC grafts. In conclusion, the BNPD-Ce6@Plt-mediated SDT is a practicable and efficacious anti-GBM therapy. Its therapeutic mechanism critically involves a synergy of tumor cell ferroptosis, macrophage stimulation, and platelet activation induced by the SDA.
Collapse
Affiliation(s)
- Meng-Fei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Jie Guo
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Shen-Jun Yuan
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Ke Li
- Center for Lab Teaching, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Quan Zhang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Hui-Mei Lei
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Jia-Lin Wu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Li Zhao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RADX), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yong-Hong Xu
- Institute of Ophthalmological Research, Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Xiao Chen
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430072, China.
| |
Collapse
|
6
|
Su W, Wang H, Pan J, Zhou Q. Advances in Sonodynamic Therapy: Focus on Ferroptosis. J Med Chem 2025; 68:5976-5992. [PMID: 40063557 DOI: 10.1021/acs.jmedchem.4c02603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Ferroptosis is a nonapoptotic form of cell death discovered in 2012. Noninvasive treatments regulating ferroptosis are important for a wide range of diseases. Among the noninvasive treatments, sonodynamic therapy (SDT) has become promising due to its strong tissue penetration and few side effects. In recent years, targeted drug delivery platforms constructed on the basis of SDT have provided an efficient delivery mode for the regulation of ferroptosis. Based on the latest research reports, this Perspective introduces the basic mechanism of SDT and the influencing factors of therapeutic effects, elucidates the significance of ferroptosis-targeted SDT, and summarizes the recent studies on ferroptosis-targeted SDT through different pathways. We also present innovative studies of composite ultrasound-responsive drug delivery platforms. Finally, a brief summary and outlook based on current ferroptosis-targeted SDT are presented.
Collapse
Affiliation(s)
- Wendi Su
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hao Wang
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Juhong Pan
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qing Zhou
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
7
|
Kang K, Wu Y, Zhang X, Wang S, Ni S, Shao J, Du Y, Yu Y, Shen Y, Chen Y, Chen W. An endoplasmic reticulum and lipid droplets dual-localized strategy to develop small molecular photosensitizers that induce ferroptosis during photodynamic therapy. Eur J Med Chem 2025; 286:117306. [PMID: 39854940 DOI: 10.1016/j.ejmech.2025.117306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/13/2025] [Accepted: 01/19/2025] [Indexed: 01/27/2025]
Abstract
Organelle-localized photosensitizers have been well-developed to enhance the photodynamic therapy (PDT) efficacy through triggering given cell death. The endoplasmic reticulum (ER) and lipid droplets (LDs) are two key organelles mutually regulating ferroptosis. Thus, in this study, small molecular photosensitizer CAR PSs were developed through fragment integration strategy and the heavy-atom modification. It was showed that the integration strategy did not affect the organelle localization and CAR PSs successfully achieved ER/LDs dual location. Besides, the heavy-atom modification help CAR PSs display good ROS generation efficiency. Importantly, ER/LDs dual-localized CAR PSs exhibited superior photo-toxicity and lower dark-toxicity against multiple breast cancer cell lines than the only ER-targeting Ce6, which further explained the superposition effect of dual organelle targeting. Preliminary studies revealed that CAR PSs induced enhanced ferroptosis via simultaneously triggering the ER stress and lipid peroxidation during PDT. Moreover, CAR-2 demonstrated significant in vivo PDT activity to suppress the tumor growth in 4T1 tumor bearing mice. These findings not only provide a promising photosensitizer CAR-2 exerting excellent in vitro and in vivo PDT effect through stimulating ferroptosis, but also propose a design strategy for the development of ER/LDs dual localized PSs.
Collapse
Affiliation(s)
- Ke Kang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - You Wu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Xi Zhang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang, 321299, China
| | - Shuqi Wang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Shaokai Ni
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Jiaan Shao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, 310015, China
| | - Yushen Du
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Yongping Yu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang, 321299, China
| | - Yong Shen
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China.
| | - Yiding Chen
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China.
| | - Wenteng Chen
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang, 321299, China.
| |
Collapse
|
8
|
Xiong K, Luo G, Zeng W, Wen G, Wang C, Ding A, Qi M, Liu Y, Zhang J. Magnetic Microbubbles Combined with ICG-Loaded Liposomes for Synergistic Mild-Photothermal and Ferroptosis-Enhanced Photodynamic Therapy of Melanoma. Int J Nanomedicine 2025; 20:2901-2921. [PMID: 40093542 PMCID: PMC11908402 DOI: 10.2147/ijn.s503753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
Background Melanoma poses a significant threat to human health due to the lack of effective treatment options. Previous studies have demonstrated that the combination of photothermal therapy (PTT) and photodynamic therapy (PDT) can enhance therapeutic efficacy. However, conventional PTT/PDT combination strategies face various challenges, including complex preparation processes, potential damage to healthy tissues, and insufficient generation of reactive oxygen species (ROS). This study aims to design a rational and efficient PTT/PDT therapeutic strategy for melanoma and to explore its underlying mechanisms. Methods We first synthesized two target materials, indocyanine green-targeted liposomes (ICG-Lips) and magnetic microbubbles (MMBs), using the thin-film hydration method, followed by characterization and performance evaluation of both materials. Subsequently, we evaluated the synergistic therapeutic effects and underlying mechanisms of ICG-Lips combined with MMBs in melanoma treatment through in vitro experiments using cellular models and in vivo experiments using animal models. Results Herein, we developed a multifunctional system comprising ICG-Lips and MMBs. ICG-Lips enhance targeted delivery through specific binding to the S100B protein on melanoma cells, while MMBs, via ultrasound (US)-induced cavitation effects, shorten the uptake time of ICG-Lips by melanoma cells and improve uptake efficiency. Furthermore, the combination of ICG-Lips and MMBs induces significant reactive oxygen species (ROS) generation. Under 808 nm laser irradiation, the accumulation of ICG-Lips in melanoma cells achieves mild photothermal therapy (mPTT) and PDT effects. The elevated temperature and excessive ROS generated during these processes result in glutathione (GSH) depletion, ultimately triggering ferroptosis. The occurrence of ferroptosis further amplifies PDT efficacy, creating a synergistic effect that effectively suppresses melanoma growth. Additionally, the combined therapeutic strategy of ICG-Lips and MMBs demonstrates excellent biosafety. Conclusion In summary, this study presents a novel and straightforward strategy that integrates mPTT, PDT, and ferroptosis synergistically to combat melanoma, thereby laying a solid foundation for improving melanoma treatment outcomes.
Collapse
Affiliation(s)
- Kaifen Xiong
- Department of Dermatology, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Guanghong Luo
- Department of Radiation Oncology, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Wei Zeng
- Department of Ultrasonography, Shenzhen People’s Hospital, second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Guanxi Wen
- Department of Ultrasonography, Shenzhen People’s Hospital, second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Chong Wang
- Department of Dermatology, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, Guangdong, People’s Republic of China
- Department of Geriatrics, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Aijia Ding
- Department of Dermatology, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Min Qi
- Department of Plastic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, People’s Republic of China
| | - Yingying Liu
- Department of Ultrasonography, Shenzhen People’s Hospital, second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Jianglin Zhang
- Department of Dermatology, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, Guangdong, People’s Republic of China
- Department of Geriatrics, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
9
|
Xu W, Guan G, Yue R, Dong Z, Lei L, Kang H, Song G. Chemical Design of Magnetic Nanomaterials for Imaging and Ferroptosis-Based Cancer Therapy. Chem Rev 2025; 125:1897-1961. [PMID: 39951340 DOI: 10.1021/acs.chemrev.4c00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Ferroptosis, an iron-dependent form of regulatory cell death, has garnered significant interest as a therapeutic target in cancer treatment due to its distinct characteristics, including lipid peroxide generation and redox imbalance. However, its clinical application in oncology is currently limited by issues such as suboptimal efficacy and potential off-target effects. The advent of nanotechnology has provided a new way for overcoming these challenges through the development of activatable magnetic nanoparticles (MNPs). These innovative MNPs are designed to improve the specificity and efficacy of ferroptosis induction. This Review delves into the chemical and biological principles guiding the design of MNPs for ferroptosis-based cancer therapies and imaging-guided therapies. It discusses the regulatory mechanisms and biological attributes of ferroptosis, the chemical composition of MNPs, their mechanism of action as ferroptosis inducers, and their integration with advanced imaging techniques for therapeutic monitoring. Additionally, we examine the convergence of ferroptosis with other therapeutic strategies, including chemodynamic therapy, photothermal therapy, photodynamic therapy, sonodynamic therapy, and immunotherapy, within the context of nanomedicine strategies utilizing MNPs. This Review highlights the potential of these multifunctional MNPs to surpass the limitations of conventional treatments, envisioning a future of drug-resistance-free, precision diagnostics and ferroptosis-based therapies for treating recalcitrant cancers.
Collapse
Affiliation(s)
- Wei Xu
- School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, PR China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Guoqiang Guan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Renye Yue
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, PR China
| | - Zhe Dong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Lingling Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Heemin Kang
- Department of Materials Science and Engineering and College of Medicine, Korea University, 12 Seoul 02841, Republic of Korea
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| |
Collapse
|
10
|
Li W, Lin Z, Liu J, Zhang J, Li Y, Liu Y, Yuan X, Li H, Shen H. Pt(IV) prodrug as a potent nanosonosensitizer self-cyclically amplifies sonodynamic-chemotherapy with dually reversing cisplatin resistance. J Mater Chem B 2025; 13:3186-3197. [PMID: 39905853 DOI: 10.1039/d4tb02615b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Although sonodynamic therapy (SDT) has shown promising advancements in combination with chemotherapy, it frequently necessitates the requirement of conventional sonosensitizers and chemotherapeutic agents, engendering intricate systems and potential drug resistance. Herein, we fabricated a potent Pt(IV)-poly(amino acid) coordination nanosonosensitizer (PHPt) with dual reversal of cisplatin resistance, producing abundant 1O2 and ˙OH upon ultrasound irradiation without the use of any external sonosensitizers. The Pt(IV) prodrug in PHPt efficiently reduced to cisplatin through SDT-induced ˙H and glutathione (GSH), inducing ˙OH accumulation and CDDP release, which further amplified the oxidative stress on SDT. Moreover, the high GSH depletion performance of PHPt and administration of aspirin effectively inhibited cisplatin detoxification and activation of the nuclear factor-kappa B pathway, respectively. This cooperative action between the Pt(IV) prodrug and SDT in the tumor microenvironment promoted self-cyclic amplification of sonodynamic-chemotherapy, achieving a significant tumor inhibition rate of 99.4%. Thus, this study offers novel perspectives on the sonosensitizer development and cisplatin application in SDT.
Collapse
Affiliation(s)
- Wenxin Li
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Ziyi Lin
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Jiahui Liu
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Jiarui Zhang
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Yuxuan Li
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Yian Liu
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Xinru Yuan
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Huimin Li
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Heyun Shen
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
11
|
Lu X, Wang Z, Zhang Y, Meng T, Chen X, Yuan R, Liu B, He H, Ding X, Zhang S. A curcumin-based HDACs inhibitor for targeted sonodynamic therapy of breast cancer. Int J Biol Macromol 2025; 287:138616. [PMID: 39672420 DOI: 10.1016/j.ijbiomac.2024.138616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024]
Abstract
Histone Deacetylases (HDACs) have emerged as key therapeutic targets in cancer treatment. In this study, we designed CURSAHA, a multifunctional anticancer agent, through the pharmacophore fusion of Vorinostat and curcumin. CURSAHA demonstrates broad-spectrum inhibitory activity against HDACs, effectively suppressing tumor cells with overexpressed HDACs. Notably, CURSAHA generates reactive oxygen species (ROS) under ultrasonic conditions, exhibiting sonodynamic therapeutic activity. Additionally, CURSAHA downregulates HDACs through redox reactions involving ROS. These properties enable CURSAHA to exhibit robust antitumor activity in both in vitro and in vivo models, highlighting its potential as a promising candidate for further development in cancer therapy.
Collapse
Affiliation(s)
- Xing Lu
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou Engineering Laboratory for Synthetic Drugs, School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Ziwei Wang
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou Engineering Laboratory for Synthetic Drugs, School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China; College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China
| | - Yu Zhang
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou Engineering Laboratory for Synthetic Drugs, School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Ti Meng
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou Engineering Laboratory for Synthetic Drugs, School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Xuehua Chen
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou Engineering Laboratory for Synthetic Drugs, School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Rongmiao Yuan
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou Engineering Laboratory for Synthetic Drugs, School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Bing Liu
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou Engineering Laboratory for Synthetic Drugs, School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Huan He
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou Engineering Laboratory for Synthetic Drugs, School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China; College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China.
| | - Xin Ding
- Hubei Key Laboratory of Pollutant Analysis & Reuse Technology, Hubei Normal University, Huangshi 435002, PR China.
| | - Silong Zhang
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou Engineering Laboratory for Synthetic Drugs, School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China; College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China.
| |
Collapse
|
12
|
Wu H, Cheng K, Sun Z, Lu Y, Guo Q, Li C. Trends of mapping knowledge structure and themes of cancer sonodynamic therapy: a text-mining study. Quant Imaging Med Surg 2024; 14:8734-8757. [PMID: 39698701 PMCID: PMC11651988 DOI: 10.21037/qims-24-128] [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: 01/20/2024] [Accepted: 09/14/2024] [Indexed: 12/20/2024]
Abstract
Background Sonodynamic therapy (SDT) is a non-invasive cancer treatment technique stemming from photodynamic therapy (PDT) and has garnered escalated interest among researchers in recent years. Numerous aspects of cancer SDT remain contentious, and the global research trajectory within this domain remains insufficiently explored. This study seeks to delineate the comprehensive knowledge framework, developmental trends, and pivotal research focal points concerning cancer SDT. Methods The study retrieved documents on cancer SDT from the Web of Science Core Collection (WoSCC) database spanning from 1 January 2000 to 7 December 2023. Bibliometric visualization was carried out through the utilization of CiteSpace 6.2 R6, VOSviewer 1.6.20, and an online analytical platform. Several bibliometric techniques including co-authorship, co-citation, co-occurrence, cluster, as well as burst analysis were used. Results A total of 672 publications including 603 articles and 69 reviews were included. The annual publication count exhibited a steady increase over time, notably experiencing a surge, particularly in recent years. In terms of contributors, China has maintained its prominent position with the highest outputs and the most financial support. Chinese Academy of Sciences contributed the most articles. Materials Science was the most investigated research areas. Breast cancer emerged as the most extensively studied tumor, succeeded by sarcoma, hepatocellular carcinoma, melanoma, pancreatic cancer, glioma. According to co-cited references, "harnessing nanomaterial", "sonodynamic precision tumor therapy" and "metal-organic framework" denote the current and emerging research focuses within the field. In tandem with the results from keywords co-occurrence and burst, we identified the following research topics including mechanism of induced cell death (ferroptosis, immunogenic cell death), nano-related research (nanoplatform, nanozymes, nanomaterials, nanosheets, metal-organic frameworks (MOFs), nanocomposites, nanoparticles, nanosonosensitizers, liposomes, nanocarriers), combination therapies (chemodynamic therapy, immunotherapy, radiotherapy, photothermal therapy), and tumor microenvironment (hypoxia, singlet oxygen, oxidative stress), that may remain the research hotspots and receive sustained attention in the near future. Conclusions For the first time, this bibliometric analysis not only presents a comprehensive portrayal of the knowledge framework, but also delineates shifts in research focal points related to cancer SDT within the last two decades. This systematic summarization offers a comprehensive and lucid comprehension of cancer SDT, providing valuable insights for further investigations in this domain.
Collapse
Affiliation(s)
- Haiyang Wu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Kunming Cheng
- Department of Intensive Care Unit, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zaijie Sun
- Department of Orthopaedic Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Yanqiu Lu
- Department of Intensive Care Unit, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiang Guo
- Department of Spine and Joint Surgery, Tianjin Medical University Baodi Hospital, Tianjin, China
| | - Cheng Li
- Department of Spine Surgery, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Center for Musculoskeletal Surgery (CMSC), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt University of Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
13
|
Zeng YL, Liu LY, Ma TZ, Liu Y, Liu B, Liu W, Shen QH, Wu C, Mao ZW. Iridium(III) Photosensitizers Induce Simultaneous Pyroptosis and Ferroptosis for Multi-Network Synergistic Tumor Immunotherapy. Angew Chem Int Ed Engl 2024; 63:e202410803. [PMID: 39180126 DOI: 10.1002/anie.202410803] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 08/26/2024]
Abstract
The integration of pyroptosis and ferroptosis hybrid cell death induction to augment immune activation represents a promising avenue for anti-tumor treatment, but there is a lack of research. Herein, we developed two iridium (III)-triphenylamine photosensitizers, IrC and IrF, with the capacity to disrupt redox balance and induce photo-driven cascade damage to DNA and Kelch-like ECH-associated protein 1 (KEAP1). The activation of the absent in melanoma 2 (AIM2)-related cytoplasmic nucleic acid-sensing pathway, triggered by damaged DNA, leads to the induction of gasdermin D (GSDMD)-mediated pyroptosis. Simultaneously, iron homeostasis, regulated by the KEAP1/nuclear factor erythroid 2-related factor 2 (NRF2)/heme oxygenase 1 (HO-1) pathway, serves as a pivotal bridge, facilitating not only the induction of gasdermin E (GSDME)-mediated non-canonical pyroptosis, but also ferroptosis in synergy with glutathione peroxidase 4 (GPX4) depletion. The collaborative action of pyroptosis and ferroptosis generates a synergistic effect that elicits immunogenic cell death, stimulates a robust immune response and effectively inhibits tumor growth in vivo. Our work introduces the first metal-based small molecule dual-inducers of pyroptosis and ferroptosis for potent cancer immunotherapy, and highlights the significance of iron homeostasis as a vital hub connecting synergistic effects of pyroptosis and ferroptosis.
Collapse
Affiliation(s)
- You-Liang Zeng
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Anti-Infective Drug Development, IGCME, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Liu-Yi Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Anti-Infective Drug Development, IGCME, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Tian-Zhu Ma
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Anti-Infective Drug Development, IGCME, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Yu Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Anti-Infective Drug Development, IGCME, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Bin Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Anti-Infective Drug Development, IGCME, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Wenting Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Anti-Infective Drug Development, IGCME, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Qing-Hua Shen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Anti-Infective Drug Development, IGCME, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Chao Wu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, 510080, P. R. China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Anti-Infective Drug Development, IGCME, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| |
Collapse
|
14
|
Huang D, Yao Y, Lou Y, Kou L, Yao Q, Chen R. Disulfiram and cancer immunotherapy: Advanced nano-delivery systems and potential therapeutic strategies. Int J Pharm X 2024; 8:100307. [PMID: 39678262 PMCID: PMC11638648 DOI: 10.1016/j.ijpx.2024.100307] [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: 08/22/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
The initial focus of the clinical application of disulfiram was its efficacy in treating alcoholism. However, recent research has revealed its potential as an anti-tumor agent and even as an enhancer of cancer immunotherapy. Disulfiram has received safety approval from the FDA, indicating its safety advantages over other substances used for disease treatment. Although clinical trials have been conducted on strategies involving disulfiram or its combination with other anti-tumor drugs, the treatment outcomes have not yielded satisfactory results, thereby emphasizing the significance of addressing drug delivery as a crucial challenge to be resolved. The need to explore advanced nano-delivery systems and the potential immunotherapy enhancement effect of disulfiram in cancer treatment has increased. This review highlights various ways in which disulfiram can combat cancer and importantly, activate immune-related mechanisms. It also discusses obstacles related to delivering disulfiram and provides existing solutions in terms of drug delivery. These drug delivery strategies offer solutions to address various challenges encountered in diverse delivery methods and aim to achieve enhanced therapeutic effects. The focus is on recent advancements in disulfiram delivery strategies and the future potential of disulfiram in immune regulation.
Collapse
Affiliation(s)
- Di Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Yinsha Yao
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Yifei Lou
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
15
|
Ge A, Xiang W, Li Y, Zhao D, Chen J, Daga P, Dai CC, Yang K, Yan Y, Hao M, Zhang B, Xiao W. Broadening horizons: the multifaceted role of ferroptosis in breast cancer. Front Immunol 2024; 15:1455741. [PMID: 39664391 PMCID: PMC11631881 DOI: 10.3389/fimmu.2024.1455741] [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: 06/27/2024] [Accepted: 10/21/2024] [Indexed: 12/13/2024] Open
Abstract
Breast cancer poses a serious threat to women's health globally. Current radiotherapy and chemotherapy regimens can induce drug-resistance effects in cancer tissues, such as anti-apoptosis, anti-pyroptosis, and anti-necroptosis, leading to poor clinical outcomes in the treatment of breast cancer. Ferroptosis is a novel programmed cell death modality characterized by iron overload, excessive generation of reactive oxygen species, and membrane lipid peroxidation. The occurrence of ferroptosis results from the imbalance between intracellular peroxidation mechanisms (executive system) and antioxidant mechanisms (defensive system), specifically involving iron metabolism pathways, amino acid metabolism pathways, and lipid metabolism pathways. In recent years, it has been found that ferroptosis is associated with the progression of various diseases, including tumors, hypertension, diabetes, and Alzheimer's disease. Studies have confirmed that triggering ferroptosis in breast cancer cells can significantly inhibit cancer cell proliferation and invasion, and improve cancer cell sensitivity to radiotherapy and chemotherapy, making induction of ferroptosis a potential strategy for the treatment of breast cancer. This paper reviews the development of the concept of ferroptosis, the mechanisms of ferroptosis (including signaling pathways such as GSH-GPX4, FSP1-CoQ1, DHODH-CoQ10, and GCH1-BH4) in breast cancer disease, the latest research progress, and summarizes the research on ferroptosis in breast cancer disease within the framework of metabolism, reactive oxygen biology, and iron biology. The key regulatory factors and mechanisms of ferroptosis in breast cancer disease, as well as important concepts and significant open questions in the field of ferroptosis and related natural compounds, are introduced. It is hoped that future research will make further breakthroughs in the regulatory mechanisms of ferroptosis and the use of ferroptosis in treating breast cancer cells. Meanwhile, natural compounds may also become a new direction for potential drug development targeting ferroptosis in breast cancer treatment. This provides a theoretical basis and opens up a new pathway for research and the development of drugs for the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wang Xiang
- Department of Rheumatology, The First People’s Hospital Changde City, Changde, Hunan, China
| | - Yan Li
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Da Zhao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Junpeng Chen
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, United States
- Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China
| | - Pawan Daga
- Department of Internal Medicine, University of Louisville, Louisville, KY, United States
| | - Charles C. Dai
- Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States
| | - Kailin Yang
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yexing Yan
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Moujia Hao
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | | | - Wei Xiao
- Department of Rheumatology, The First People’s Hospital Changde City, Changde, Hunan, China
| |
Collapse
|
16
|
Han B, Liu Y, Zhou Q, Yu Y, Liu X, Guo Y, Zheng X, Zhou M, Yu H, Wang W. The advance of ultrasound-enabled diagnostics and therapeutics. J Control Release 2024; 375:1-19. [PMID: 39208935 DOI: 10.1016/j.jconrel.2024.08.039] [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: 04/30/2024] [Revised: 07/27/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Point-of-care ultrasound demonstrates significant potential in biomedical research due to its noninvasive, real-time visualization, cost-effectiveness, and other biological benefits. Ultrasound irradiation can precisely control the mechanical and physicochemical effects on pathogenic lesions, enabling real-time visualization, tunable tissue penetration depth, and therapeutic applications. This review summarizes recent advancements in ultrasound-enabled diagnostics and therapeutics, focusing on mechanochemical effects that can be directly integrated into biomedical applications. Additionally, the structure-functionality relationships of sonotheranostic nanoplatforms are systematically discussed, providing insights into the underlying biological effects. Finally, the limitations of current ultrasonic medicine are discussed, along with potential expansions to facilitate patient-centered translations.
Collapse
Affiliation(s)
- Biying Han
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yan Liu
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Qianqian Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yuting Yu
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Xingxing Liu
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yu Guo
- State Key Laboratory of Chemical Biology & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaohua Zheng
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Mengjiao Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Haijun Yu
- State Key Laboratory of Chemical Biology & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
17
|
Yang F, Lv J, Ma W, Yang Y, Hu X, Yang Z. Engineering Sonosensitizer-Derived Nanotheranostics for Augmented Sonodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402669. [PMID: 38970544 DOI: 10.1002/smll.202402669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/13/2024] [Indexed: 07/08/2024]
Abstract
Sonodynamic therapy (SDT), featuring noninvasive, deeper penetration, low cost, and repeatability, is a promising therapy approach for deep-seated tumors. However, the general or only utilization of SDT shows low efficiency and unsatisfactory treatment outcomes due to the complicated tumor microenvironment (TME) and SDT process. To circumvent the issues, three feasible approaches for enhancing SDT-based therapeutic effects, including sonosensitizer optimization, strategies for conquering hypoxia TME, and combinational therapy are summarized, with a particular focus on the combination therapy of SDT with other therapy modalities, including chemodynamic therapy, photodynamic therapy, photothermal therapy, chemotherapy, starvation therapy, gas therapy, and immunotherapy. In the end, the current challenges in SDT-based therapy on tumors are discussed and feasible approaches for enhanced therapeutic effects are provided. It is envisioned that this review will provide new insight into the strategic design of high-efficiency sonosensitizer-derived nanotheranostics, thereby augmenting SDT and accelerating the potential clinical transformation.
Collapse
Affiliation(s)
- Fuhong Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Jingqi Lv
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Wen Ma
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Yanling Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Xiaoming Hu
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
- Jiangxi Key Laboratory of Nanobiomaterials, School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, China
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| |
Collapse
|
18
|
Wen D, Feng J, Deng R, Li K, Zhang H. Zn/Pt dual-site single-atom driven difunctional superimposition-augmented sonosensitizer for sonodynamic therapy boosted ferroptosis of cancer. Nat Commun 2024; 15:9359. [PMID: 39472589 PMCID: PMC11522694 DOI: 10.1038/s41467-024-53488-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/14/2024] [Indexed: 11/02/2024] Open
Abstract
Sonodynamic therapy (SDT) as a non-invasive antitumor strategy has been widely concerned. However, the rapid electron (e-) and hole (h+) recombination of traditional inorganic semiconductor sonosensitizers under ultrasonic (US) stimulation greatly limits the production of reactive oxygen species (ROS). Herein, we report a unique Zn/Pt dual-site single-atom driven difunctional superimposition-augmented TiO2-based sonosensitizer (Zn/Pt SATs). Initially, we verify through theoretical calculation that the strongly coupled Zn and Pt atoms can assist electron excitation at the atomic level by increasing electron conductivity and excitation efficiency under US, respectively, thus effectively improving the yield of ROS. Additionally, Zn/Pt SATs can significantly enhance ferroptosis by producing more ROS and sonoexcited holes under US stimuli. Therefore, the establishment of dual-site single-atom system represents an innovative strategy to enhance SDT in cancer model of female mice and provides a typical example for the development of inorganic sonosensitizer in the field of antitumor therapy.
Collapse
Affiliation(s)
- Ding Wen
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China
- University of Science and Technology of China, 230026, Hefei, China
| | - Jing Feng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China.
- University of Science and Technology of China, 230026, Hefei, China.
| | - Ruiping Deng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China
| | - Kai Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China.
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China.
- University of Science and Technology of China, 230026, Hefei, China.
- Department of Chemistry, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
19
|
Wang H, Li D, Wang H, Ren Q, Pan Y, Dao A, Wang D, Wang Z, Zhang P, Huang H. Enhanced Sonodynamic Therapy for Deep Tumors Using a Self-Assembled Organoplatinum(II) Sonosensitizer. J Med Chem 2024; 67:18356-18367. [PMID: 39360515 DOI: 10.1021/acs.jmedchem.4c01671] [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: 10/04/2024]
Abstract
Despite the promising advances in photodynamic therapy (PDT), it remains challenging to target and treat deep-seated solid tumors effectively. Herein, we developed an organoplatinum(II) complex (Pt-TPE) with self-assembly properties for sonodynamic therapy (SDT). Pt-TPE forms a nanofiber network structure through Pt-Pt and π-π stacking interactions. Notably, under ultrasound (US), Pt-TPE demonstrates unique self-assembly-induced singlet oxygen (1O2) generation due to a significantly enhanced singlet-triplet intersystem crossing (ISC). This generation of 1O2 occurs exclusively in the self-assembled state of Pt-TPE. Additionally, Pt-TPE exhibits sono-cytotoxicity against cancer cells by impairing mitochondrial membrane potential (MMP), inhibiting glucose uptake, and aerobic glycolysis. Furthermore, US-activated Pt-TPE significantly inhibits deep solid tumors in mice, achieving remarkable therapeutic efficacy even at penetration depths greater than 10 cm. This study highlights the potential of self-assembled metal complexes to enhance the efficacy of SDT for treating deep tumors.
Collapse
Affiliation(s)
- Haobing Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Dan Li
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Hanqiang Wang
- Department of Chemistry and Dongguan Key Laboratory for Data Science and Intelligent Medicine, Great Bay University, Dongguan 523000, China
| | - Qingyan Ren
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Anyi Dao
- School of Pharmaceutical Science, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Deliang Wang
- Department of Materials Chemistry, Huzhou University, Huzhou 313000, China
| | - Zhigang Wang
- School of Pharmacy, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Pingyu Zhang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Huaiyi Huang
- School of Pharmaceutical Science, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| |
Collapse
|
20
|
Li Y, Chang P, Xu L, Zhu Z, Hu M, Cen J, Li S, Zhao YE. TiO2-Nanoparticle-Enhanced Sonodynamic Therapy for Prevention of Posterior Capsular Opacification and Ferroptosis Exploration of Its Mechanism. Invest Ophthalmol Vis Sci 2024; 65:24. [PMID: 39417751 PMCID: PMC11500051 DOI: 10.1167/iovs.65.12.24] [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: 03/05/2024] [Accepted: 07/30/2024] [Indexed: 10/19/2024] Open
Abstract
Purpose To explore the application and potential ferroptosis mechanisms of sonodynamic therapy (SDT) using titanium dioxide nanoparticles (TiO2-NPs) as sonosensitizers for the prevention of posterior capsule opacification (PCO). Methods We fabricated TiO2-NP-coated intraocular lenses (TiO2-IOLs) using the spin-coating method, followed by ultrasound activation of the photosensitizer TiO2. In vitro experiments were performed with human lens epithelial cells (HLECs) to explore the appropriate concentration of TiO2 and ultrasonic parameters. Investigations included reactive oxygen species (ROS) generation, glutathione (GSH) depletion, glutathione peroxidase 4 (GPX4) western blot analysis, lipid peroxidation assays, and transcriptomics analysis. Finally, TiO2-IOLs were implanted in rabbit eyes to explore the in vivo performance of SDT. Results Through both in vitro and in vivo experiments, the study determined that the ultrasound parameters of 5-minute duration, 1-MHz frequency, 50% duty cycle, and 1.2-W/cm2 intensity were reliable and valid for killing HLECs without damaging other ocular structures. In vitro experiments demonstrated that SDT generated excess ROS, which disrupted the mitochondrial membrane potential and significantly reduced the GSH content. Additionally, the downregulation of GPX4, accumulation of lipid peroxides, and alteration of mitochondrial morphology were observed, suggesting that ferroptosis may be the underlying mechanism. The RNA-sequencing analysis results also showed an increase in the expression of multiple pro-ferroptosis genes and the ferroptosis marker gene PTGS2. Animal experiments preliminarily demonstrated the safety and effectiveness of SDT in treating PCO in vivo. Conclusions TiO2-IOLs combined with SDT effectively prevented PCO by generating ROS and intracellular ferroptosis.
Collapse
Affiliation(s)
- Yuanyuan Li
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Pingjun Chang
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Liming Xu
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Zehui Zhu
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Man Hu
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Jiaying Cen
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Siyan Li
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Yun-e Zhao
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| |
Collapse
|
21
|
He M, Ma Z, Zhang L, Zhao Z, Zhang Z, Liu W, Wang R, Fan J, Peng X, Sun W. Sonoinduced Tumor Therapy and Metastasis Inhibition by a Ruthenium Complex with Dual Action: Superoxide Anion Sensitization and Ligand Fracture. J Am Chem Soc 2024; 146:25764-25779. [PMID: 39110478 DOI: 10.1021/jacs.4c08278] [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: 09/19/2024]
Abstract
Photoresponsive ruthenium(II) complexes have recently emerged as a promising tool for synergistic photodynamic therapy and chemotherapy in oncology, as well as for antimicrobial applications. However, the limited penetration power of photons prevents the treatment of deep-seated lesions. In this study, we introduce a sonoresponsive ruthenium complex capable of generating superoxide anion (O2•-) via type I process and initiating a ligand fracture process upon ultrasound triggering. Attaching hydroxyflavone (HF) as an "electron reservoir" to the octahedral-polypyridyl-ruthenium complex resulted in decreased highest occupied molecular orbital (HOMO)-lowest unoccupied molecular orbital (LUMO) energy gaps and triplet-state metal to ligand charge transfer (3MLCT) state energy (0.89 eV). This modification enhanced the generation of O2•- under therapeutic ultrasound irradiation at a frequency of 1 MHz. The produced O2•- rapidly induced an intramolecular cascade reaction and HF ligand fracture. As a proof-of-concept, we engineered the Ru complex into a metallopolymer platform (PolyRuHF), which could be activated by low-power ultrasound (1.5 W cm-2, 1.0 MHz, 50% duty cycle) within a centimeter range of tissue. This activation led to O2•- generation and the release of cytotoxic ruthenium complexes. Consequently, PolyRuHF induced cellular apoptosis and ferroptosis by causing mitochondrial dysfunction and excessive toxic lipid peroxidation. Furthermore, PolyRuHF effectively inhibited subcutaneous and orthotopic breast tumors and prevented lung metastasis by downregulating metastasis-related proteins in mice. This study introduces the first sonoresponsive ruthenium complex for sonodynamic therapy/sonoactivated chemotherapy, offering new avenues for deep tumor treatment.
Collapse
Affiliation(s)
- Maomao He
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Zhiyuan Ma
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Linhao Zhang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Zhiyu Zhao
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Zongwei Zhang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Wenkai Liu
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Ran Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
22
|
Wang Y, Meng L, Zhao F, Zhao L, Gao W, Yu Q, Chen P, Sun Y. Harnessing External Irradiation for Precise Activation of Metal-Based Agents in Cancer Therapy. Chembiochem 2024; 25:e202400305. [PMID: 38825577 DOI: 10.1002/cbic.202400305] [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: 04/03/2024] [Revised: 06/01/2024] [Accepted: 06/02/2024] [Indexed: 06/04/2024]
Abstract
Cancer is a significant global health issue. Platinum-based chemotherapy drugs, including cisplatin, are crucial in clinical anti-cancer treatment. However, these drugs have limitations such as drug resistance, non-specific distribution, and irreversible toxic and side effects. In recent years, the development of metal-based agents has led to the discovery of other anti-cancer effects beyond chemotherapy. Precise spatiotemporal controlled external irradiation can activate metal-based agents at specific sites and play a different role from traditional chemotherapy. These strategies can not only enhance the anti-cancer efficiency, but also show fewer side effects and non-cross-drug resistance, which are ideal approaches to solve the problems caused by traditional platinum-based chemotherapy drugs. In this review, we focus on various metal-based agent-mediated cancer therapies that are activated by three types of external irradiation: near-infrared (NIR) light, ultrasound (US), and X-ray, and give some prospects. We hope that this review will promote the generation of new kinds of metal-based anti-cancer agents.
Collapse
Affiliation(s)
- Yuting Wang
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Liling Meng
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Fang Zhao
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, 430079, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, 530021, China
| | - Limei Zhao
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Wei Gao
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Qi Yu
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Peiyao Chen
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Yao Sun
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| |
Collapse
|
23
|
Lupica-Tondo GL, Arner EN, Mogilenko DA, Voss K. Immunometabolism of ferroptosis in the tumor microenvironment. Front Oncol 2024; 14:1441338. [PMID: 39188677 PMCID: PMC11345167 DOI: 10.3389/fonc.2024.1441338] [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: 05/30/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
Ferroptosis is an iron-dependent form of cell death that results from excess lipid peroxidation in cellular membranes. Within the last decade, physiological and pathological roles for ferroptosis have been uncovered in autoimmune diseases, inflammatory conditions, infection, and cancer biology. Excitingly, cancer cell metabolism may be targeted to induce death by ferroptosis in cancers that are resistant to other forms of cell death. Ferroptosis sensitivity is regulated by oxidative stress, lipid metabolism, and iron metabolism, which are all influenced by the tumor microenvironment (TME). Whereas some cancer cell types have been shown to adapt to these stressors, it is not clear how immune cells regulate their sensitivities to ferroptosis. In this review, we discuss the mechanisms of ferroptosis sensitivity in different immune cell subsets, how ferroptosis influences which immune cells infiltrate the TME, and how these interactions can determine epithelial-to-mesenchymal transition (EMT) and metastasis. While much focus has been placed on inducing ferroptosis in cancer cells, these are important considerations for how ferroptosis-modulating strategies impact anti-tumor immunity. From this perspective, we also discuss some promising immunotherapies in the field of ferroptosis and the challenges associated with targeting ferroptosis in specific immune cell populations.
Collapse
Affiliation(s)
- Gian Luca Lupica-Tondo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Emily N. Arner
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Denis A. Mogilenko
- Department of Medicine, Department of Pathology, Microbiology and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kelsey Voss
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
24
|
Zhao J, Bian E, Zhang R, Xu T, Nie Y, Wang L, Jin G, Xie H, Xiang H, Chen Y, Wu D. Self-Assembled Aza-Boron-Dipyrromethene-Based H 2S Prodrug for Synergistic Ferroptosis-Enabled Gas and Sonodynamic Tumor Therapies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309542. [PMID: 38872263 PMCID: PMC11321684 DOI: 10.1002/advs.202309542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/12/2024] [Indexed: 06/15/2024]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and lethal subtype of gliomas of the central nervous system. The efficacy of sonodynamic therapy (SDT) against GBM is significantly reduced by the expression of apoptosis-inhibitory proteins in GBM cells. In this study, an intelligent nanoplatform (denoted as Aza-BD@PC NPs) based on the aza-boron-dipyrromethene dye and phenyl chlorothionocarbonate-modified DSPE-PEG molecules is developed for synergistic ferroptosis-enabled gas therapy (GT) and SDT of GBM. Once internalized by GBM cells, Aza-BD@PC NPs showed effective cysteine (Cys) consumption and Cys-triggered hydrogen sulfide (H2S) release for ferroptosis-enabled GT, thereby disrupting homeostasis in the intracellular environment, affecting GBM cell metabolism, and inhibiting GBM cell proliferation. Additionally, the released Aza-BD generated abundant singlet oxygen (1O2) under ultrasound irradiation for favorable SDT. In vivo and in vitro evaluations demonstrated that the combined functions of Cys consumption, H2S production, and 1O2 production induced significant death of GBM cells and markedly inhibited tumor growth, with an impressive inhibition rate of up to 97.5%. Collectively, this study constructed a cascade nanoreactor with satisfactory Cys depletion performance, excellent H2S release capability, and prominent reactive oxygen species production ability under ultrasound irradiation for the synergistic ferroptosis-enabled GT and SDT of gliomas.
Collapse
Affiliation(s)
- Jiajia Zhao
- Department of NeurosurgeryThe Second Affiliated Hospital of Anhui Medical UniversityHefei230601P. R. China
| | - Erbao Bian
- Department of NeurosurgeryThe Second Affiliated Hospital of Anhui Medical UniversityHefei230601P. R. China
| | - Renwu Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital of Anhui Medical UniversityHefei230601P. R. China
| | - Tao Xu
- Department of NeurosurgeryChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Yang Nie
- Department of NeurosurgeryThe Second Affiliated Hospital of Anhui Medical UniversityHefei230601P. R. China
| | - Linqi Wang
- Department of NeurosurgeryThe Second Affiliated Hospital of Anhui Medical UniversityHefei230601P. R. China
| | - Gui Jin
- Department of NeurosurgeryThe Second Affiliated Hospital of Anhui Medical UniversityHefei230601P. R. China
| | - Han Xie
- Department of NeurosurgeryThe Second Affiliated Hospital of Anhui Medical UniversityHefei230601P. R. China
| | - Huijing Xiang
- School of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Yu Chen
- School of Life SciencesShanghai UniversityShanghai200444P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou Institute of Shanghai UniversityWenzhou325088P. R. China
- Shanghai Institute of MaterdicineShanghai200051P. R. China
| | - Dejun Wu
- Department of NeurosurgeryThe Second Affiliated Hospital of Anhui Medical UniversityHefei230601P. R. China
| |
Collapse
|
25
|
Li D, Fan M, Wang H, Zhu Y, Yu B, Zhang P, Huang H. Facile synthesis of a hydrazone-based zinc(ii) complex for ferroptosis-augmented sonodynamic therapy. Chem Sci 2024; 15:10027-10035. [PMID: 38966369 PMCID: PMC11220576 DOI: 10.1039/d4sc02102a] [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: 03/30/2024] [Accepted: 05/07/2024] [Indexed: 07/06/2024] Open
Abstract
Sonodynamic therapy (SDT), as a novel non-invasive cancer treatment modality derived from photodynamic therapy (PDT), has drawn much attention due to its unique advantages for the treatment of deep tumors. Zinc-based complexes have shown great clinical prospect in PDT due to their excellent photodynamic activity and biosafety. However, their application in SDT has lagged seriously behind. Exploring efficient zinc-based complexes as sono-sensitizers remains an appealing but significantly challenging task. Herein, we develop a hydrazone ligand-based zinc complex (ZnAMTC) for SDT of tumors in vitro and in vivo. ZnAMTC was facilely synthesized via a two-step reaction from low-cost raw materials without tedious purification. It shows negligible dark toxicity and can produce singlet oxygen (1O2) under ultrasound (US) irradiation, exhibiting high sono-cytotoxicity to various cancer cells. Mechanism studies show that ZnAMTC can effectively reduce the levels of glutathione (GSH) and glutathione peroxidase 4 (GPX4) under US irradiation and later cause ferroptosis of cancer cells. In vivo studies further demonstrate that ZnAMTC exhibits efficient tumor growth inhibition under US irradiation and has good biosafety. This work provides useful insights into the design of first-row transition metal complexes for SDT application.
Collapse
Affiliation(s)
- Dan Li
- College of Chemistry and Environmental Engineering, Shenzhen University Shenzhen 518060 China
| | - Minghui Fan
- College of Chemistry and Environmental Engineering, Shenzhen University Shenzhen 518060 China
| | - Haobing Wang
- College of Chemistry and Environmental Engineering, Shenzhen University Shenzhen 518060 China
| | - Yongjie Zhu
- College of Chemistry and Environmental Engineering, Shenzhen University Shenzhen 518060 China
| | - Bole Yu
- Laboratory of Life Science, Shenzhen Research Institute of the Hong Kong Polytechnic University Shenzhen 518057 China
| | - Pingyu Zhang
- College of Chemistry and Environmental Engineering, Shenzhen University Shenzhen 518060 China
| | - Huaiyi Huang
- School of Pharmaceutical Science (Shenzhen), Shenzhen Campus of Sun Yat-sen University No. 66, Gongchang Road Shenzhen 518107 China
| |
Collapse
|
26
|
Luo Y, Bai XY, Zhang L, Hu QQ, Zhang N, Cheng JZ, Hou MZ, Liu XL. Ferroptosis in Cancer Therapy: Mechanisms, Small Molecule Inducers, and Novel Approaches. Drug Des Devel Ther 2024; 18:2485-2529. [PMID: 38919962 PMCID: PMC11198730 DOI: 10.2147/dddt.s472178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
Ferroptosis, a unique form of programmed cell death, is initiated by an excess of iron accumulation and lipid peroxidation-induced damage. There is a growing body of evidence indicating that ferroptosis plays a critical role in the advancement of tumors. The increased metabolic activity and higher iron levels in tumor cells make them particularly vulnerable to ferroptosis. As a result, the targeted induction of ferroptosis is becoming an increasingly promising approach for cancer treatment. This review offers an overview of the regulatory mechanisms of ferroptosis, delves into the mechanism of action of traditional small molecule ferroptosis inducers and their effects on various tumors. In addition, the latest progress in inducing ferroptosis using new means such as proteolysis-targeting chimeras (PROTACs), photodynamic therapy (PDT), sonodynamic therapy (SDT) and nanomaterials is summarized. Finally, this review discusses the challenges and opportunities in the development of ferroptosis-inducing agents, focusing on discovering new targets, improving selectivity, and reducing toxic and side effects.
Collapse
Affiliation(s)
- YiLin Luo
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Xin Yue Bai
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Lei Zhang
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Qian Qian Hu
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Ning Zhang
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Jun Zhi Cheng
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Ming Zheng Hou
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Xiao Long Liu
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| |
Collapse
|
27
|
Wu J, Huang J, Yu J, Xu M, Liu J, Pu K. Exosome-Inhibiting Polymeric Sonosensitizer for Tumor-Specific Sonodynamic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400762. [PMID: 38445783 DOI: 10.1002/adma.202400762] [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: 01/15/2024] [Revised: 03/04/2024] [Indexed: 03/07/2024]
Abstract
Combination cancer immunotherapy based on electromagnetic energy and immunotherapy shows potent anti-cancer efficacy. However, as a factor that mediates tumor metastasis and immune suppression, the impact of tumor exosomes on therapy under electromagnetic energy stimulation remains unclear. Herein, findings indicate that sonodynamic therapy (SDT) increases serum exosome levels by inducing apoptotic exosomes and loosening the tumor extracellular matrix, promoting lung metastasis. To address this problem, an exosome-inhibiting polymeric sonosensitizer (EIPS) selectively inhibiting tumor exosome generation in response to the tumor biomarker is synthesized. EIPS consists of a semiconducting polymer backbone capable of inducing SDT and a poly(ethylene glycol) layer conjugated with a tumor-specific enzyme-responsive exosome inhibitor prodrug. After being cleaved by tumor Cathepsin B, EIPS releases active exosome inhibitors, preventing tumor exosome-mediated immune suppression and lung metastasis. As a result, EIPS elicits robust antitumor effects through the synergistic effect of SDT and tumor exosome inhibition, completely preventing lung metastasis and establishing a long-term immune memory effect. This is the first example showing that combining SDT with tumor-specific exosome inhibition can elicit a potent immune response without the help of typical immune agonists.
Collapse
Affiliation(s)
- Jiayan Wu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jingsheng Huang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jie Yu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
| | - Mengke Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| |
Collapse
|
28
|
Kushwaha R, Singh V, Peters S, Yadav AK, Sadhukhan T, Koch B, Banerjee S. Comparative Study of Sonodynamic and Photoactivated Cancer Therapies with Re(I)-Tricarbonyl Complexes Comprising Phenanthroline Ligands. J Med Chem 2024; 67:6537-6548. [PMID: 38603561 DOI: 10.1021/acs.jmedchem.3c02485] [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: 04/13/2024]
Abstract
Herein, we have compared the effectivity of light-based photoactivated cancer therapy and ultrasound-based sonodynamic therapy with Re(I)-tricarbonyl complexes (Re1-Re3) against cancer cells. The observed photophysical and TD-DFT calculations indicated the potential of Re1-Re3 to act as good anticancer agents under visible light/ultrasound exposure. Re1 did not display any dark- or light- or ultrasound-triggered anticancer activity. However, Re2 and Re3 displayed concentration-dependent anticancer activity upon light and ultrasound exposure. Interestingly, Re3 produced 1O2 and OH• on light/ultrasound exposure. Moreover, Re3 induced NADH photo-oxidation in PBS and produced H2O2. To the best of our knowledge, NADH photo-oxidation has been achieved here with the Re(I) complex for the first time in PBS. Additionally, Re3 released CO upon light/ultrasound exposure. The cell death mechanism revealed that Re3 produced an apoptotic cell death response in HeLa cells via ROS generation. Interestingly, Re3 showed slightly better anticancer activity under light exposure compared to ultrasound exposure.
Collapse
Affiliation(s)
- Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Virendra Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Silda Peters
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Ashish Kumar Yadav
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Tumpa Sadhukhan
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Biplob Koch
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
29
|
Huang Y, Ouyang W, Lai Z, Qiu G, Bu Z, Zhu X, Wang Q, Yu Y, Liu J. Nanotechnology-enabled sonodynamic therapy against malignant tumors. NANOSCALE ADVANCES 2024; 6:1974-1991. [PMID: 38633037 PMCID: PMC11019498 DOI: 10.1039/d3na00738c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/09/2024] [Indexed: 04/19/2024]
Abstract
Sonodynamic therapy (SDT) is an emerging approach for malignant tumor treatment, offering high precision, deep tissue penetration, and minimal side effects. The rapid advancements in nanotechnology, particularly in cancer treatment, have enhanced the efficacy and targeting specificity of SDT. Combining sonodynamic therapy with nanotechnology offers a promising direction for future cancer treatments. In this review, we first systematically discussed the anti-tumor mechanism of SDT and then summarized the common nanotechnology-related sonosensitizers and their recent applications. Subsequently, nanotechnology-related therapies derived using the SDT mechanism were elaborated. Finally, the role of nanomaterials in SDT combined therapy was also introduced.
Collapse
Affiliation(s)
- Yunxi Huang
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| | - Wenhao Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Yat-sen Supercomputer Intelligent Medical Joint Research Institute, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University 510120 Guangzhou China
| | - Zijia Lai
- First Clinical Medical College, Guangdong Medical University 524000 Zhanjiang China
| | - Guanhua Qiu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| | - Zhaoting Bu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| | - Xiaoqi Zhu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| | - Qin Wang
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| | - Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Yat-sen Supercomputer Intelligent Medical Joint Research Institute, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University 510120 Guangzhou China
- Faculty of Medicine, Macau University of Science and Technology Taipa Macao PR China
| | - Junjie Liu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| |
Collapse
|
30
|
Zhang B, Huang Y, Huang Y. Advances in Nanodynamic Therapy for Cancer Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:648. [PMID: 38607182 PMCID: PMC11013863 DOI: 10.3390/nano14070648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/04/2024] [Accepted: 04/07/2024] [Indexed: 04/13/2024]
Abstract
Nanodynamic therapy (NDT) exerts its anti-tumor effect by activating nanosensitizers to generate large amounts of reactive oxygen species (ROS) in tumor cells. NDT enhances tumor-specific targeting and selectivity by leveraging the tumor microenvironment (TME) and mechanisms that boost anti-tumor immune responses. It also minimizes damage to surrounding healthy tissues and enhances cytotoxicity in tumor cells, showing promise in cancer treatment, with significant potential. This review covers the research progress in five major nanodynamic therapies: photodynamic therapy (PDT), electrodynamic therapy (EDT), sonodynamic therapy (SDT), radiodynamic therapy (RDT), and chemodynamic therapy (CDT), emphasizing the significant role of advanced nanotechnology in the development of NDT for anti-tumor purposes. The mechanisms, effects, and challenges faced by these NDTs are discussed, along with their respective solutions for enhancing anti-tumor efficacy, such as pH response, oxygen delivery, and combined immunotherapy. Finally, this review briefly addresses challenges in the clinical translation of NDT.
Collapse
Affiliation(s)
| | | | - Yong Huang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China; (B.Z.); (Y.H.)
| |
Collapse
|
31
|
Guo C, Geng HJ, Wang WJ, Liu YX, Deng L, Tian JM, Gao JM, Tang JJ. Dimerized sesquiterpenoid [4 + 2] adducts with ferroptosis-promoting activity from Inula britannica Linn. PHYTOCHEMISTRY 2024; 218:113951. [PMID: 38096962 DOI: 10.1016/j.phytochem.2023.113951] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/29/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Inubritanolides C and D (1 and 2), two exo sesquiterpenoid [4 + 2] adducts with unprecedented interconverting conformations of twist-chair and chair, together with two previously undescribed endo [4 + 2] dimers (3 and 4) were discovered from Inula britannica flowers. Dimers 1 and 2 have an undescribed carbon skeleton comprising of eudesmanolide and guaianolide units with the linkage mode of C-11/C-1' and C-13/C-3' via a Diels-Alder cycloaddition reaction. Their structures were elucidated using 1D/2D NMR, X-ray diffraction, ECD, and variable-temperature NMR experiments. Dimer 2 displayed a strong inhibitory effect on breast cancer cells by promoting lipid ROS production, showing its potential as ferroptosis inducer.
Collapse
Affiliation(s)
- Cong Guo
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Hui-Jun Geng
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Wen-Ji Wang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Yan-Xiang Liu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Lu Deng
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Jun-Mian Tian
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China.
| | - Jin-Ming Gao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Jiang-Jiang Tang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
32
|
Zhou YC, Zhao TK, Tao SM, Wang P, Guan YC, Yang KP, Chen SQ, Pu XY. Recent Progress in Ferroptosis Induced Tumor Cell Death by Anti-tumor Metallic complexes. Chem Asian J 2024; 19:e202301020. [PMID: 38149729 DOI: 10.1002/asia.202301020] [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: 11/19/2023] [Revised: 12/21/2023] [Accepted: 12/25/2023] [Indexed: 12/28/2023]
Abstract
Metal complexes represented by platinum complexes play a very important role in cancer treatment due to their diverse chemical structures and anti-tumor activities. Recently, ferroptosis has emerged as a newly occurring cell death form in the anti-tumor process. It has been reported that metal complexes could inhibit the proliferation and metastasis of tumors and combat chemotherapy resistance by targeting ferroptosis. In this review, we briefly describe ferroptosis as a fundamental process for tumor suppression and triggering anti-tumor immune responses. We summarize recent developments on metal complexes that induce ferroptosis. Finally, we outline the prospects for the application of metal complexes to the treatment of tumors based on ferroptosis and the associated problems that need to be solved, and discussed other potential research directions of metal complexes.
Collapse
Affiliation(s)
- Yong-Chang Zhou
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Tian-Kun Zhao
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Si-Man Tao
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Peng Wang
- School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Yi-Chen Guan
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Ke-Pei Yang
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Sheng-Qiang Chen
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Xiu-Ying Pu
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| |
Collapse
|
33
|
Liang CJ, Wu RC, Huang XQ, Qin QP, Liang H, Tan MX. Synthesis and anticancer mechanisms of four novel platinum(II) 4'-substituted-2,2':6',2''-terpyridine complexes. Dalton Trans 2024; 53:2143-2152. [PMID: 38189098 DOI: 10.1039/d3dt03197g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Mitophagy, a selective autophagic process, has emerged as a pathway involved in degrading dysfunctional mitochondria. Herein, new platinum(II)-based chemotherapeutics with mitophagy-targeting properties are proposed. Four novel binuclear anticancer Pt(II) complexes with 4'-substituted-2,2':6',2''-terpyridine derivatives (tpy1-tpy4), i.e., [Pt2(tpy1)(DMSO)2Cl4]·CH3OH (tpy1Pt), [Pt(tpy2)Cl][Pt(DMSO)Cl3]·CH3COCH3 (tpy2Pt), [Pt(tpy3)Cl][Pt(DMSO)Cl3] (tpy3Pt), and [Pt(tpy4)Cl]Cl·CH3OH (tpy4Pt), were designed and prepared. Moreover, their potential antitumor mechanism was studied. Tpy1Pt-tpy4Pt exhibited more selective cytotoxicity against cisplatin-resistant SK-OV-3/DDP (SKO3cisR) cancer cells compared with those against ovarian SK-OV-3 (SKO3) cancer cells and normal HL-7702 liver (H702) cells. This selective cytotoxicity of Tpy1Pt-tpy4Pt was better than that of its ligands (i.e., tpy1-tpy4), the clinical drug cisplatin, and cis-Pt(DMSO)2Cl2. The results of various experiments indicated that tpy1Pt and tpy2Pt kill SKO3cisR cancer cells via a mitophagy pathway, which involves the disruption of the mitophagy-related protein expression, dissipation of the mitochondrial membrane potential, elevation of the [Ca2+] and reactive oxygen species levels, promotion of mitochondrial DNA damage, and reduction in the adenosine triphosphate and mitochondrial respiratory chain levels. Furthermore, in vivo experiments indicated that the dinuclear anticancer Pt(II) coordination compound (tpy1Pt) has remarkable therapeutic efficiency (ca. 52.4%) and almost no toxicity. Therefore, the new 4'-substituted-2,2':6',2''-terpyridine Pt(II) coordination compound (tpy1Pt) is a potential candidate for next-generation mitophagy-targeting dinuclear Pt(II)-based anticancer drugs.
Collapse
Affiliation(s)
- Chun-Jie Liang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Run-Chun Wu
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Xiao-Qiong Huang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Qi-Pin Qin
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China
| | - Ming-Xiong Tan
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| |
Collapse
|
34
|
Wang H, Lai Y, Li D, Karges J, Zhang P, Huang H. Self-Assembly of Erlotinib-Platinum(II) Complexes for Epidermal Growth Factor Receptor-Targeted Photodynamic Therapy. J Med Chem 2024; 67:1336-1346. [PMID: 38183413 DOI: 10.1021/acs.jmedchem.3c01889] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2024]
Abstract
Due to cell mutation and self-adaptation, the application of clinical drugs with early epidermal growth factor receptor (EGFR)-targeted inhibitors is severely limited. To overcome this limitation, herein, the synthesis and in-depth biological evaluation of an erlotinib-platinum(II) complex as an EGFR-targeted anticancer agent is reported. The metal complex is able to self-assemble inside an aqueous solution and readily form nanostructures with strong photophysical properties. While being poorly toxic toward healthy cells and upon treatment in the dark, the compound was able to induce a cytotoxic effect in the very low micromolar range upon irradiation against EGFR overexpressing (drug resistant) human lung cancer cells as well as multicellular tumor spheroids. Mechanistic insights revealed that the compound was able to selectively degrade the EGFR using the lysosomal degradation pathway upon generation of singlet oxygen at the EGFR. We are confident that this work will open new avenues for the treatment of EGFR-overexpressing tumors.
Collapse
Affiliation(s)
- Haobing Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yidan Lai
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
- School of Pharmaceutical Science, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Dan Li
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Bochum 44780, Germany
| | - Pingyu Zhang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Huaiyi Huang
- School of Pharmaceutical Science, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
35
|
Li X, Sun X, Chen H, Chen X, Li Y, Li D, Zhang Z, Chen H, Gao Y. Exploring BODIPY derivatives as sonosensitizers for anticancer sonodynamic therapy. Eur J Med Chem 2024; 264:116035. [PMID: 38101040 DOI: 10.1016/j.ejmech.2023.116035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/23/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023]
Abstract
Sonodynamic therapy (SDT) is an emerging non-invasive and effective therapeutic modality for cancer treatment bearing benefit of deep tissue-penetration in comparison to photo-inspired therapy. However, exploring novel sonosensitizers with high sonosensitivity and desirable biosafety remains a significant challenge. Although boron dipyrromethene (BODIPY) dyes have been widely used in biomedical filed, no BODIPY-based sonosensitizers have been reported yet. Herein, we synthesized four BODIPY dyes (BDP1-BDP4) and investigated their potential applications in SDT. BDP4 exhibited superb sonosensitivity and high SDT efficiency against cancer cells and tumors in tumor-bearing mice. The types of the generated reactive oxygen species, cavitation effect, and cell apoptosis were investigated to figure out the sonodynamic therapeutic mechanisms of BDP4. This work for the first time demonstrates the potential of BODIPY dyes as novel sonosensitizers for SDT, which may pave an avenue for developing more efficient and safer sonosensitizers in future.
Collapse
Affiliation(s)
- Xudong Li
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350108, China
| | - Xianbin Sun
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350108, China
| | - Hui Chen
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Xinyu Chen
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350108, China
| | - Yuanming Li
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350108, China
| | - Dongmiao Li
- State Key Lab of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Zizhong Zhang
- State Key Lab of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Haijun Chen
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China.
| | - Yu Gao
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350108, China.
| |
Collapse
|
36
|
He F, Li W, Liu B, Zhong Y, Jin Q, Qin X. Progress of Piezoelectric Semiconductor Nanomaterials in Sonodynamic Cancer Therapy. ACS Biomater Sci Eng 2024; 10:298-312. [PMID: 38124374 DOI: 10.1021/acsbiomaterials.3c01232] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Sonodynamic therapy is an emerging noninvasive tumor treatment method that utilizes ultrasound to stimulate sonosensitizers to produce a large amount of reactive oxygen species, inducing tumor cell death. Though sonodynamic therapy has very promising prospects in cancer treatment, the application of early organic sonosensitizers has been limited in efficacy due to the high blood clearance-rate, poor water solubility, and low stability. Inorganic sonosensitizers have thus been developed, among which piezoelectric semiconductor materials have received increasing attention in sonodynamic therapy due to their piezoelectric properties and strong stability. In this review, we summarized the designs, principles, modification strategies, and applications of several commonly used piezoelectric materials in sonodynamic therapy and prospected the future clinical applications for piezoelectric semiconductor materials in sonodynamic therapy.
Collapse
Affiliation(s)
- Fang He
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Clinical Research Center for Medical Imaging in Hubei Province, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Wenqu Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Clinical Research Center for Medical Imaging in Hubei Province, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Beibei Liu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Clinical Research Center for Medical Imaging in Hubei Province, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yi Zhong
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Clinical Research Center for Medical Imaging in Hubei Province, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Clinical Research Center for Medical Imaging in Hubei Province, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Xiaojuan Qin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Clinical Research Center for Medical Imaging in Hubei Province, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 1277 Jiefang Avenue, Wuhan 430022, China
| |
Collapse
|
37
|
Sun T, Wang R, Lu W, Shi X, Gao F, Wu T, Wang G, Su X, Teng Z. Platinum nanoparticle-anchored metal-organic complex nanospheres by a coordination-crystallization approach for enhanced sonodynamic therapy of tumors. J Mater Chem B 2023; 11:11280-11289. [PMID: 37990931 DOI: 10.1039/d3tb02497k] [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: 11/23/2023]
Abstract
The combination of noble metal nanoparticles with metal-organic complexes has attracted great attention for exploring new properties in biomedical application areas. So far, the preparation of noble metal nanoparticle-loaded metal-organic complexes often requires complex processes. Here, a simple coordination-crystallization approach was developed to prepare platinum nanoparticle-anchored metal-organic complexes (Pt-MOCs) by directly mixing disulfiram (DSF), chloroplatinic acid, and a reducing agent. The DSF and Pt ions first coordinate forming metal-organic complex nanospheres and then the Pt nanoparticles crystallized on the surface taking advantage of the coordination rate of the metal ions and organic ligand being greater than the reduction rate of the metal ions. The Pt-MOCs possess uniform and adjustable diameter (240-536 nm), and their surface potentials can also be modulated easily from -22 to +14 mV by adjusting the ratio of DSF and chloroplatinic acid. Phantom experiments show that the Pt-MOC nanospheres significantly improve the efficiency of singlet oxygen production after exposure to ultrasound irradiation. In vitro experiments show that the Pt-MOCs effectively produce reactive oxygen species and exhibit superior cytotoxicity for tumor cells under ultrasound irradiation compared to metal-organic complexes (MOCs) or Pt nanoparticles. Taken together, this work reports a coordination-crystallization approach to synthesize Pt-MOCs, which show excellent sonodynamic therapy for tumors.
Collapse
Affiliation(s)
- Tangyao Sun
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Rui Wang
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Wei Lu
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Xuzhi Shi
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Shandong 250012, P. R. China.
| | - Tingting Wu
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Guoqin Wang
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Xiaodan Su
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| |
Collapse
|
38
|
Lv M, Zheng Y, Wu J, Shen Z, Guo B, Hu G, Huang Y, Zhao J, Qian Y, Su Z, Wu C, Xue X, Liu HK, Mao ZW. Evoking Ferroptosis by Synergistic Enhancement of a Cyclopentadienyl Iridium-Betulin Immune Agonist. Angew Chem Int Ed Engl 2023; 62:e202312897. [PMID: 37830171 DOI: 10.1002/anie.202312897] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/07/2023] [Accepted: 10/12/2023] [Indexed: 10/14/2023]
Abstract
Ferroptosis is a form of programmed cell death driven by iron-dependent lipid peroxidation (LPO) with the potential for antitumor immunity activation. In this study, a nonferrous cyclopentadienyl metal-based ferroptosis inducer [Ir(Cp*)(Bet)Cl]Cl (Ir-Bet) was developed by a metal-ligand synergistic enhancement (MLSE) strategy involving the reaction of [Ir(Cp*)Cl]2 Cl2 with the natural product Betulin. The fusion of Betulin with iridium cyclopentadienyl (Ir-Cp*) species as Ir-Bet not only tremendously enhanced the antiproliferative activity toward cancer cells, but also activated ferritinophagy for iron homeostasis regulation by PI3K/Akt/mTOR cascade inhibition with a lower dosage of Betulin, and then evoked an immune response by nuclear factor kappa-B (NF-κB) activation of Ir-Cp* species. Further immunogenic cell death (ICD) occurred by remarkable ferroptosis through glutathione (GSH) depletion, glutathione peroxidase 4 (GPX4) deactivation and ferritinophagy. An in vivo vaccination experiment demonstrated desirable antitumor and immunogenic effects of Ir-Bet by increasing the ratio of cytotoxic T cells (CTLs)/regulatory T cells (Tregs).
Collapse
Affiliation(s)
- Mengdi Lv
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Yue Zheng
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, IGCME, GBRCE for Functional Molecular Engineering, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Jian Wu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Zhengqi Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Binglian Guo
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Guojing Hu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Yuanlei Huang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Jingyue Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Yong Qian
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Zhi Su
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Chao Wu
- Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Xuling Xue
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Hong-Ke Liu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, IGCME, GBRCE for Functional Molecular Engineering, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| |
Collapse
|
39
|
Sun W, Xiao H, Zhu J, Hao Z, Sun J, Wang D, Wang X, Ramalingam M, Xie S, Wang R. Multifunctional Oxygen-Generating Nanoflowers for Enhanced Tumor Therapy. ACS APPLIED BIO MATERIALS 2023; 6:4998-5008. [PMID: 37880964 DOI: 10.1021/acsabm.3c00678] [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: 10/27/2023]
Abstract
Sonodynamic therapy (SDT) and chemotherapy have received great attention as effective methods for tumor treatment. However, the inherent hypoxia of the tumor greatly hinders its therapeutic efficacy. In this work, a tumor microenvironment-responsive biodegradable nanoplatform SiO2-MnO2-PEG-Ce6&DOX (designated as SMPC&D) is fabricated by encapsulating manganese oxide (MnO2) into silica nanoparticles and anchoring poly(ethylene glycol) (PEG) onto the surface for tumor hypoxia relief and delivery, then loaded with sonosensitizer Chlorin e6 (Ce6) and chemotherapeutic drug doxorubicin (DOX) for hypoxic tumor treatment. We evaluated the physicochemical properties of SMPC&D nanoparticles and the tumor therapeutic effects of chemotherapy and SDT under ultrasound stimulation in vitro and in vivo. After endocytosis by tumor cells, highly expressed glutathione (GSH) triggers biodegradation of the nanoplatform and MnO2 catalyzes hydrogen peroxide (H2O2) to generate oxygen (O2), thereby alleviating tumor hypoxia. Depleting GSH and self-supplying O2 effectively improve the SDT efficiency both in vitro and in vivo. Ultrasonic stimulation promoted the release and cellular uptake of chemotherapy drugs. In addition, the relieved hypoxia reduced the efflux of chemotherapy drugs by downregulating the expression of the P-gp protein, which jointly improved the effect of chemotherapy. This study demonstrates that the degradable SMPC&D as a therapeutic agent can achieve efficient chemotherapy and SDT synergistic therapy for hypoxic tumors.
Collapse
Affiliation(s)
- Wanru Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
- Science Fund of Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yantai 264000, People's Republic of China
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Huifang Xiao
- Zhongnan Hospital of Wuhan University, Wuhan 430062, People's Republic of China
| | - Jiazhi Zhu
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Zhaokun Hao
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Jian Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Deqiang Wang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264100, People's Republic of China
| | - Xin Wang
- Department of Rehabilitation Medicine, Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Murugan Ramalingam
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country(UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
- Joint Research Laboratory (JRL), Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Drug Formulation Unit 10, Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
- Bioprinting and Precisión Medicine, Centro de investigación Lascaray Ikergunea, Avenida Miguel de Unamuno, 01006 Vitoria-Gasteiz, Spain
- School of Basic Medical Science, Chengdu University, Chengdu 610106, China
- Department of Metallurgical and Materials Engineering, Atilim University, Ankara 06830, Turkey
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, 78054 Villingen-Schwennigen, Germany
| | - Shuyang Xie
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Ranran Wang
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
- Science Fund of Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yantai 264000, People's Republic of China
| |
Collapse
|
40
|
Wang X, Wang C, Tian H, Chen Y, Wu B, Cheng W. IR-820@NBs Combined with MG-132 Enhances the Anti-Hepatocellular Carcinoma Effect of Sonodynamic Therapy. Int J Nanomedicine 2023; 18:6199-6212. [PMID: 37933299 PMCID: PMC10625775 DOI: 10.2147/ijn.s431910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/23/2023] [Indexed: 11/08/2023] Open
Abstract
Purpose Sonodynamic therapy (SDT) is a promising and significant measure for the treatment of tumors. However, the internal situation of hepatocellular carcinoma (HCC) is complex, separate SDT treatment is difficult to play a good therapeutic effect. Here, we used SDT combined with MG-132 to mediate apoptosis and autophagy of HCC cells to achieve the purpose of treatment of cancer. Methods To determine the generated reactive oxygen species (ROS) and the change of mitochondrial membrane potential (ΔΨm), HepG2 cells were stained by 2,7-dichlorodihydrofluorescein diacetate (DCFH-DA) and 5,5',6,6'-Tetrachloro-1,1',3,3'-tetraethyl-imidacarbocyanine iodide (JC-1) staining to determine the IR-820@NBs-mediated SDT to achieve HCC therapy through the mitochondrial pathway. Cell counting kit 8 (CCK-8) assay and flow cytometry were used to detect cell viability and apoptosis rate of HepG2 cells. Autophagy was detected by mCherry-GFP-LC3B fluorescence labeling. Chloroquine (Cq) pretreatment was used to explore the relationship between autophagy and apoptosis. To detect the ability of HepG2 cells migration and invasion, cell scratch assay and transwell assay were used. Results The successfully prepared IR-820@NBs could effectively overcome the shortcomings of IR-820 and induce lethal levels of ROS by ultrasound irradiation. As a dual agonist of apoptosis and autophagy, MG-132 could effectively enhance the efficacy of SDT in the process of treating HCC. After pre-treatment with Cq, the cell activity increased and the level of apoptosis decreased, which proved that apoptosis and autophagy were induced by combined therapy, autophagy, and apoptosis have the synergistic anti-tumor effect, and part of apoptosis was autophagy-dependent. After combined therapy, the activity and invasive ability of HCC cells decreased significantly. Conclusion SDT combined with MG-132 in the process of treating liver cancer could effectively induce apoptosis and autophagy anti-tumor therapy, which is helpful to the research of new methods to treat liver cancer.
Collapse
Affiliation(s)
- Xiaodong Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Chunyue Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Huimin Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yichi Chen
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Bolin Wu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| |
Collapse
|
41
|
Ding X, Wang Z, Yu Q, Michał N, Roman S, Liu Y, Peng N. Superoxide Dismutase-Like Regulated Fe/Ppa@PDA/B for Synergistically Targeting Ferroptosis/Apoptosis to Enhance Anti-Tumor Efficacy. Adv Healthc Mater 2023; 12:e2301824. [PMID: 37485811 DOI: 10.1002/adhm.202301824] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/06/2023] [Indexed: 07/25/2023]
Abstract
The cell apoptosis pathway of sonodynamic therapy (SDT) is usually blocked, resulting in limited therapeutic efficacy, therefore, the development of new methods for sensitizing targeted ferroptosis and promoting apoptosis is of great significance to improve the anti-tumor efficacy of SDT. Herein, mesoporous Fe3 O4 nanoparticles (NPs) are synthesized for loading pyropheophorbide-a (ppa), surface-coated by polydopamine (PDA) and further anchored with tumor-targeting moieties of biotin to obtain Fe/ppa@PDA/B NPs. Fe/ppa@PDA/B displayes pH/ultrasound (US) responsive release properties, and magnetic resonance imaging (MRI) functions. Moreover, Fe3 O4 NPs of Fe/ppa@PDA/B as the Fe source for ferroptosis, enhances ferroptosis sensitivity by consuming glutathione (GSH) and producing hydroxyl radical (OH). The quinone groups of PDA layer on Fe/ppa@PDA/B own free electrons, which led to effective superoxide dismutase (SOD) action through superoxide anion (O2 - ) disproportionation to hydrogen peroxide (H2 O2 ) and oxygen (O2 ), thus, overcame hypoxia of SDT and promoted ·OH generation by Fe ions under US trigger, synergistically improves ferroptosis and apoptosis to enhance the anti-tumor efficacy of SDT both in vitro and in vivo. The anti-tumor strategy of synergistic apoptosis and ferroptosis induce by GSH depletion and self-sufficient O2 regulated by SOD provides a new idea for enhancing SDT efficacy.
Collapse
Affiliation(s)
- Xin Ding
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Materials and Nanotechnology, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, 430081, P. R. China
- Hubei Key Laboratory of Pollutant Analysis & Reuse Technology, College of Chemistry and Chemical Engineering, Hubei Normal University, Huangshi, 435002, P. R. China
| | - Zidong Wang
- Hubei Key Laboratory of Pollutant Analysis & Reuse Technology, College of Chemistry and Chemical Engineering, Hubei Normal University, Huangshi, 435002, P. R. China
| | - Qiying Yu
- Central laboratory, Tumor Hospital Affiliated to Nantong University, Nantong, 226361, P. R. China
| | - Nowicki Michał
- Institute of Metrology and Biomedical Engineering Faculty of Mechatronics, Warsaw University of Technology, Warsaw, 00-661, Poland
| | - Szewczyk Roman
- Institute of Metrology and Biomedical Engineering Faculty of Mechatronics, Warsaw University of Technology, Warsaw, 00-661, Poland
| | - Yi Liu
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Materials and Nanotechnology, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, 430081, P. R. China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan, 430023, P. R. China
- Hubei Key Laboratory of Radiation Chemistry and Functional Materials, Hubei University of Science and Technology, Xianning, 437100, P. R. China
| | - Na Peng
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Materials and Nanotechnology, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, 430081, P. R. China
- Belt and Road Joint Laboratory on Measurement and Control Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| |
Collapse
|
42
|
Mandal AA, Kushwaha R, Yadav AK, Banerjee S. Metal Complexes for Cancer Sonodynamic Therapy. Chembiochem 2023; 24:e202200597. [PMID: 36385722 DOI: 10.1002/cbic.202200597] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 11/18/2022]
Abstract
Sonodynamic therapy (SDT) for cancer treatment is gaining attention owing to its non-invasive property and ultrasound's (US) deep tissue penetration ability. In SDT, US activates the sonosensitizer at the target deep-seated tumors to generate reactive oxygen species (ROS), which ultimately damage tumors. However, drawbacks such as insufficient ROS production, aggregation of sonosensitizer, off-target side effects, etc., of the current organic/nanomaterial-based sonosensitizers limit the effectiveness of cancer SDT. Very recently, metal complexes with tunable physiochemical properties (such as sonostability, HOMO to LUMO energy gap, ROS generation ability, aqueous solubility, emission, etc.) have been devised as effective sonosensitizers, which could overcome the limitations of organic/nanomaterial-based sonosensitizers. This concept introduces all the reported metal-based sonosensitizers and delineates the prospects of metal complexes in cancer sonodynamic therapy. This new concept of metal-based sonosensitizer can deliver next-generation cancer drugs.
Collapse
|
43
|
An J, Hong H, Won M, Rha H, Ding Q, Kang N, Kang H, Kim JS. Mechanical stimuli-driven cancer therapeutics. Chem Soc Rev 2023; 52:30-46. [PMID: 36511945 DOI: 10.1039/d2cs00546h] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mechanical stimulation utilizing deep tissue-penetrating and focusable energy sources, such as ultrasound and magnetic fields, is regarded as an emerging patient-friendly and effective therapeutic strategy to overcome the limitations of conventional cancer therapies based on fundamental external stimuli such as light, heat, electricity, radiation, or microwaves. Recent efforts have suggested that mechanical stimuli-driven cancer therapy (henceforth referred to as "mechanical cancer therapy") could provide a direct therapeutic effect and intelligent control to augment other anti-cancer systems as a synergistic combinational cancer treatment. This review article highlights the latest advances in mechanical cancer therapy to present a novel perspective on the fundamental principles of ultrasound- and magnetic field-mediated mechanical forces, including compression, tension, shear force, and torque, that can be generated in a cellular microenvironment using mechanical stimuli-activated functional materials. Additionally, this article will shed light on mechanical cancer therapy and inspire future research to pursue the development of ultrasound- and magnetic-field-activated materials and their applications in this field.
Collapse
Affiliation(s)
- Jusung An
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| | - Hyunsik Hong
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea.
| | - Miae Won
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| | - Hyeonji Rha
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| | - Qihang Ding
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| | - Nayeon Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea.
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea.
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| |
Collapse
|
44
|
Zhang Z, Zhang Y, Yang M, Hu C, Liao H, Li D, Du Y. Synergistic antibacterial effects of ultrasound combined nanoparticles encapsulated with cellulase and levofloxacin on Bacillus Calmette-Guérin biofilms. Front Microbiol 2023; 14:1108064. [PMID: 36937280 PMCID: PMC10014853 DOI: 10.3389/fmicb.2023.1108064] [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: 11/25/2022] [Accepted: 01/30/2023] [Indexed: 03/06/2023] Open
Abstract
Tuberculosis is a chronic infectious disease, the treatment of which is challenging due to the formation of cellulose-containing biofilms by Mycobacterium tuberculosis (MTB). Herein, a composite nanoparticle loaded with cellulase (CL) and levofloxacin (LEV) (CL@LEV-NPs) was fabricated and then combined with ultrasound (US) irradiation to promote chemotherapy and sonodynamic antimicrobial effects on Bacillus Calmette-Guérin bacteria (BCG, a mode of MTB) biofilms. The CL@LEV-NPs containing polylactic acid-glycolic acid (PLGA) as the shell and CL and LEV as the core were encapsulated via double ultrasonic emulsification. The synthesized CL@LEV-NPs were uniformly round with an average diameter of 196.2 ± 2.89 nm, and the zeta potential of -14.96 ± 5.35 mV, displaying high biosafety and sonodynamic properties. Then, BCG biofilms were treated with ultrasound and CL@LEV-NPs separately or synergistically in vivo and in vitro. We found that ultrasound significantly promoted biofilms permeability and activated CL@LEV-NPs to generate large amounts of reactive oxygen species (ROS) in biofilms. The combined treatment of CL@LEV-NPs and US exhibited excellent anti-biofilm effects, as shown by significant reduction of biofilm biomass value and viability, destruction of biofilm architecture in vitro, elimination of biofilms from subcutaneous implant, and remission of local inflammation in vivo. Our study suggested that US combined with composite drug-loaded nanoparticles would be a novel non-invasive, safe, and effective treatment modality for the elimination of biofilm-associated infections caused by MTB.
Collapse
Affiliation(s)
- Zhifei Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Yuqing Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Min Yang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Can Hu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Hongjian Liao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Dairong Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Dairong Li,
| | - Yonghong Du
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Yonghong Du,
| |
Collapse
|