1
|
Wang B, Tang X, Xiao C, Yu Z, Bo H, Wang J, Wang J. Nucleus-targeted ruthenium(II) complex triggers immunogenic cell death and sensitizes melanoma to anti-PD-1 therapy by activating cGAS-STING pathway. J Inorg Biochem 2025; 267:112871. [PMID: 40022761 DOI: 10.1016/j.jinorgbio.2025.112871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
A significant challenge in the treatment of melanoma with immune checkpoint blockades (ICBs) is the limited T cells response often observed in immunologically "cold" tumors. By leveraging the immunogenicity of immunogenic cell death (ICD), which increases the susceptibility of tumor cells to ICBs, this study investigated the potential of a nucleus-targeted ruthenium(II) complex (Ru1) as an inducer of ICD. Treatment with Ru1 induced DNA damage in melanoma cells, activating the cyclic GMP-AMP synthase-stimulator of the interferon genes (cGAS-STING) pathway. This triggered endoplasmic reticulum (ER) stress, leading to ICD. Ru1-treated dying melanoma cells exhibited characteristics such as cell exposure of calreticulin (CRT) on the cell surface, release of adenosine triphosphate (ATP), and secretion of high-mobility group box 1 (HMGB1). Vaccination with Ru1-treated, dying melanoma cells elicited robust antitumor immune responses, as evidenced by CD8+ T cells activation, reduced Foxp3+ T cells count, and the development of a memory immune response that protected mice from subsequent melanoma challenges. Combining Ru1 with anti-PD-1 therapy significantly promoted T cells infiltration, enhanced dendritic cell activation, and reduced tumor-associated immunosuppressive factors, indicating a reprogramming of the tumor microenvironment. These findings suggest that Ru1 is a promising therapeutic agent for treating "cold" tumors in cancer chemoimmunotherapy.
Collapse
Affiliation(s)
- Bishu Wang
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Xingguo Tang
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Chuntao Xiao
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Zhijie Yu
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Huaben Bo
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Jie Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Jinquan Wang
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| |
Collapse
|
2
|
Moosavi Zenooz A, Eterafi M, Azarmi Giglou S, Safarzadeh E. Embracing cancer immunotherapy with manganese particles. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01070-9. [PMID: 40397376 DOI: 10.1007/s13402-025-01070-9] [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: 02/15/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025] Open
Abstract
A substance integral to the sustenance and functionality of virtually all forms of life is manganese (Mn), classified as an essential trace metal. Its significance lies in its pivotal role in facilitating metabolic processes crucial for survival. Additionally, Mn exerts influence over various biological functions including bone formation and maintenance, as well as regulation within systems governing immunity, nervous signaling, and digestion. Manganese nanoparticles (Mn-NP) stand out as a beacon of promise within the realm of immunotherapy, their focus honed on intricate mechanisms such as triggering immune pathways, igniting inflammasomes, inducing immunogenic cell death (ICD), and sculpting the nuances of the tumor microenvironment. These minuscule marvels have dazzled researchers with their potential in reshaping the landscape of cancer immunotherapy - serving as potent vaccine enhancers, efficient drug couriers, and formidable allies when paired with immune checkpoint inhibitors (ICIs) or cutting-edge photodynamic/photothermal therapies. Herein, we aim to provide a comprehensive review of recent advances in the application of Mn and Mn-NP in the immunotherapy of cancer. We hope that this review will display an insightful view of Mn-NPs and provide guidance for design and application of them in immune-based cancer therapies.
Collapse
Affiliation(s)
- Ali Moosavi Zenooz
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Eterafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Soheil Azarmi Giglou
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, 5166614711, Iran.
| |
Collapse
|
3
|
Bai XF, Ma JC, Zhang C, Chen Z, He J, Cheng SX, Zhang XZ. Click Chemistry-Assisted Rejuvenation of Aging T Cells Sensitizes Aged Mice to Tumor Immunotherapy. J Am Chem Soc 2025; 147:16694-16704. [PMID: 40310278 DOI: 10.1021/jacs.5c05312] [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: 05/02/2025]
Abstract
Enormous resources have been devoted to address the suboptimal response of tumor patients to immunotherapy. However, a crucial yet often overlooked factor in these effects is the strong correlation between the occurrence and development of tumors and the immune dysfunction associated with aging. Our study aims to rejuvenate aging T cells within tumor-draining lymph nodes (TdLNs) by using targeted delivery of rapamycin, a macrolide capable of mitigating aging-related decline in immune function, thereby enhancing the antitumor efficacy of immunotherapy in aged mice. The targeted delivery system relies on a bioorthogonal reaction that harnesses the click chemistry between the azide (N3) groups artificially introduced onto TdLNs and the dibenzocyclooctyne (DBCO) groups attached to the rapamycin-loaded micelles administered intradermally. Experimental data demonstrate that this approach has effectively restored the functionality of impaired CD8+ T cells in aged mice, thereby enhancing the antitumor response to immune checkpoint blockade (ICB) therapy to levels comparable to those in young mice. This study presents a promising strategy to combat the resistance to immunotherapeutic approaches commonly encountered among elderly tumor patients.
Collapse
Affiliation(s)
- Xue-Feng Bai
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Jun-Chi Ma
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Cheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Zhu Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Jinlian He
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Department of Traditional Chinese Medicine of Zhongnan Hospital, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
4
|
Wang S, Xu D, Wang Y, Zhou Y, Xiao L, Li F, Tu J, Qin W, Tian S, Zheng B, Wang Y, Yuan XL, Liu Y, Liu B. A Bifunctional Antibody Targeting PD-1 and TGF-β Signaling Has Antitumor Activity in Combination with Radiotherapy and Attenuates Radiation-Induced Lung Injury. Cancer Immunol Res 2025; 13:767-784. [PMID: 39878763 PMCID: PMC12046334 DOI: 10.1158/2326-6066.cir-23-0903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 04/03/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Radio-immunotherapy has antitumor activity but also causes toxicity, which limits its clinical application. JS-201 is a dual antibody targeting PD-1 and TGF-β signaling. We investigated the antitumor effect of JS-201 combined with radiotherapy (RT) and the effect on radiation-induced lung injury (RILI). Different tumor models were established to detect the antitumor effects of the combination of JS-201 and RT, and RILI models were established to observe the effects of JS-201. Transcriptome sequencing showed that JS-201 optimized the tumor microenvironment by inhibiting extracellular matrix formation and angiogenesis. Combining JS-201 with RT further increased the inflammatory response and immune infiltration and showed great abscopal effects in Lewis lung cancer luciferase-positive models. Single-cell sequencing demonstrated that JS-201 reduced fibroblast proliferation by inhibiting the TGF-β/Smad pathway and the release of neutrophil extracellular traps mediated by ROS, thereby relieving radiation-induced pulmonary fibrosis. In conclusion, the JS-201 and RT combination enhances antitumor effects while mitigating acute and chronic RILI, and it may have potential for translational investigation as a cancer treatment strategy.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Duo Xu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuehua Zhou
- Top Alliance Biosciences Inc., Suzhou, China
| | - Lingyan Xiao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wan Qin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sidan Tian
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Bolong Zheng
- School of Computer Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yihua Wang
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Xiang-lin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanhui Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
León-Vega II, Oregon R, Schnoor M, Vadillo E. From Ulcerative Colitis to Metastatic Colorectal Cancer: The Neutrophil Contribution. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:814-830. [PMID: 39889826 DOI: 10.1016/j.ajpath.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
Ulcerative colitis (UC) is an inflammatory colon and rectum disease affecting approximately 5 million people worldwide. There is no cure for UC, and approximately 8% of patients with UC develop colorectal cancer (CRC) by gradual acquisition of mutations driving the formation of adenomas and their progression to adenocarcinomas and metastatic disease. CRC constitutes 10% of total cancer cases worldwide and 9% of cancer deaths. Both UC and CRC have an increasing incidence worldwide. Although the epithelium has been well studied in UC and CRC, the contribution of neutrophils is less clear. Neutrophils are rapidly recruited in excessive amounts from peripheral blood to the colon during UC, and their overactivation in the proinflammatory UC tissue environment contributes to tissue damage. In CRC, the role of neutrophils is controversial, but emerging evidence suggests that their role depends on the evolution and context of the disease. The role of neutrophils in the transition from UC to CRC is even less clear. However, recent studies propose neutrophils as therapeutic targets for better clinical management of both diseases. This review summarizes the current knowledge on the roles of neutrophils in UC and CRC.
Collapse
Affiliation(s)
- Iliana I León-Vega
- Department of Molecular Biomedicine, Cinvestav-National Polytechnic Institute, Mexico City, Mexico
| | - Reyna Oregon
- Oncology Research Unit, Oncology Hospital, National Medical Center, Mexican Institute of Social Security, Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine, Cinvestav-National Polytechnic Institute, Mexico City, Mexico.
| | - Eduardo Vadillo
- Oncology Research Unit, Oncology Hospital, National Medical Center, Mexican Institute of Social Security, Mexico City, Mexico.
| |
Collapse
|
6
|
Ling Y, Liang X, Yan K, Zeng G, Zhu X, Jiang J, Lu S, Wang X, Zhou Y, Li Z, Mai W, Wang D, Chen J. Bimetallic Ca/Zn Nanoagonist Remould the Immunosuppressive Hepatocellular Carcinoma Microenvironment Following Incomplete Microwave Ablation via Pyroptosis and the STING Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500670. [PMID: 40305756 DOI: 10.1002/advs.202500670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/18/2025] [Indexed: 05/02/2025]
Abstract
During the treatment of solid tumors, local therapeutic approaches carry the risk of incomplete radical cure, which may lead to rapid tumor growth. Incomplete microwave ablation (iMWA) can induce tumors to exhibit highly invasive and uncontrollable growth, which is related to the immunosuppressive microenvironment. A multifunctional bimetallic Ca/Zn nanoagonist (PZH/Zn@CaNA) with a biomimetic liposome-modified surface to tumor tissues after iMWA is developed. In response to the acidic tumor microenvironment, the released traditional Chinese medicine preparation Pien Tze Huang (PZH) reduced protein expressions of the JAK2-STAT3 signaling pathway, thereby slowing down the proliferation and growth of hepatocellular carcinoma (HCC). Furthermore, the bimetallic ions Ca2⁺ and Zn2⁺ can cascade to enhance the killing effect of oxidative stress, generating substantial amounts of reactive oxygen species. This process induces pyroptosis and releases significant quantities of damage associated molecular patterns, thereby triggering immune activation mechanisms related to the STING pathway that reshape the immunosuppressive HCC microenvironment resulting from iMWA. This strategy markedly differs from previous chemoimmunotherapies, which not only effectively addressed the problem of conventional drugs showing heterogeneous distribution in tumor regions, but also verified the critical role played by PZH/Zn@CaNA in inhibiting iMWA-induced rapid tumor growth, regulating oxidative stress and remodeling the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Yuan Ling
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Xiayi Liang
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Kangning Yan
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Guichun Zeng
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Xiaoqi Zhu
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Jinghang Jiang
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Shaolong Lu
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Xiaobo Wang
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Yuying Zhou
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Zhaoshen Li
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Wei Mai
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Duo Wang
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Jie Chen
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| |
Collapse
|
7
|
Zhou JJ, Feng YC, Zhao ML, Guo Q, Zhao XB. Nanotechnology-driven strategies in postoperative cancer treatment: innovations in drug delivery systems. Front Pharmacol 2025; 16:1586948. [PMID: 40371327 PMCID: PMC12075547 DOI: 10.3389/fphar.2025.1586948] [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: 03/03/2025] [Accepted: 04/23/2025] [Indexed: 05/16/2025] Open
Abstract
Cancer remains a global health challenge, and this challenge comes with a significant burden. Current treatment modalities, such as surgery, chemotherapy, and radiotherapy, have their limitations. The emergence of nanomedicines presents a new frontier in postoperative cancer treatment, offering potential to inhibit tumor recurrence and manage postoperative complications. This review deeply explores the application and potential of nanomedicines in the treatment of cancer after surgery. In particular, it focuses on local drug delivery systems (LDDS), which consist of in situ injection, implantation, and spraying. LDDS can provide targeted drug delivery and controlled release, which enhancing therapeutic efficacy. At the same time, it minimizes damage to healthy tissues and reduces systemic side effects. The nanostructures of these systems are unique. They facilitate the sustained release of drugs, prolong the effects of treatment, and decrease the frequency of dosing. This is especially beneficial in the postoperative period. Despite their potential, nanomedicines have limitations. These include high production costs, concerns regarding long-term toxicity, and complex regulatory approval processes. This paper aims to analyze several aspects. These include the advantages of nanomedicines, their drug delivery systems, how they combine with multiple treatment methods, and the associated challenges. Future research should focus on certain issues. These issues are stability, tumor specificity, and clinical translation. By addressing these, the delivery methods can be optimized and their therapeutic efficacy enhanced. With the advancements in materials science and biomedical engineering, the future design of LDDS is set to become more intelligent and personalized. It will cater to the diverse needs of clinical treatment and offer hope for better outcomes in cancer patients after surgery.
Collapse
Affiliation(s)
- Jun-Jie Zhou
- The Stomatological Hospital, Anyang Sixth People’s Hospital, Anyang, China
| | | | | | | | | |
Collapse
|
8
|
Xie D, Yan X, Shang W, Ren H, Wen W, Tang BZ, Su H. Organic Radiosensitizer with Aggregation-Induced Emission Characteristics for Tumor Ablation through Synergistic Apoptosis and Immunogenic Cell Death. ACS NANO 2025; 19:14972-14986. [PMID: 40201936 DOI: 10.1021/acsnano.5c00942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Inspired by the clinical application of thermotherapy to promote the efficacy of radiotherapy, this study demonstrates the multimodal diagnostic application of pure organic nanoparticles in the combined treatment of tumors through imaging and photothermal properties. The nanoparticles developed in this study demonstrated unique properties and multiple functionalities, including excellent photostability and thermostability, strong fluorescence emission in the near-infrared-II (NIR-II) region, extremely high photothermal conversion efficiency, good biocompatibility, significant radiosensitizing properties, and effective tumor site accumulation. In vitro and in vivo evaluations demonstrated that these nanoparticles are ideal candidates for synergistic photothermal radiotherapy guided by NIR-II fluorescence, NIR-I photoacoustic, and photothermal trimodal imaging. They act as radiosensitizers by alleviating the hypoxic tumor microenvironment, modulating the cell cycle, and inducing apoptosis and immunogenic cell death during radiotherapy, which may provide a potential approach for the clinical treatment of tumors.
Collapse
Affiliation(s)
- Dalu Xie
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xueke Yan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Wenzhao Shang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hao Ren
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Wei Wen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen, Guangdong 518172, P.R. China
| | - Huifang Su
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
9
|
Wang T, Xing Z, Wang M, He C, Ma T, Wang Y, Wang X, Wu H, Li S, Cheng C, Zhao C. Bioinspired O 2-Evolution Catalysts with Proton-Coupled Electron Transfer Pathway for Portable Oxygen Generation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2502102. [PMID: 40237195 DOI: 10.1002/adma.202502102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/24/2025] [Indexed: 04/18/2025]
Abstract
Producing high-purity oxygen (O2) has a wide range of applications across diverse sectors, such as medicine, tunnel construction, the chemical industry, and fermentation. However, current O2 production methods are burdened by complexity, heavy equipment, high energy consumption, and limited adaptability to harsh environments. Here, to address this grand challenge, the de novo design of Ru-doped metal hydroxide is proposed to serve as bioinspired O2-evolution catalysts with proton-coupled electron transfer (PCET) pathway for low-energy, environmentally friendly, cost-effective, and portable O2 generation. The comprehensive studies confirm that the lattice H species in Ru-Co(OH)x-based O2-evolution catalyst can trigger a PCET pathway to optimize Ru-oxygen intermediates interactions, thus ultimately reducing reaction energy barriers and improving the activities and durabilities. Consequently, the prepared Ru-Co(OH)x-loaded membrane catalysts exhibit rapid and long-term stable O2 production capabilities. Furthermore, the proposed material design strategy of lattice H-species shows remarkable universality and adaptability to broad Ru-doped metal hydroxides. This efficient, portable, and cost-effective O2 generation technique is suggested to ensure an uninterrupted O2 supply during emergencies and in regions with limited O2 availability or air pollution, thus offering significant societal benefits in broad applications.
Collapse
Affiliation(s)
- Ting Wang
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Sichuan University, Chengdu, 610065, China
| | - Zhenyu Xing
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Sichuan University, Chengdu, 610065, China
| | - Mao Wang
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Sichuan University, Chengdu, 610065, China
| | - Chao He
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Sichuan University, Chengdu, 610065, China
| | - Tian Ma
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Sichuan University, Chengdu, 610065, China
| | - Yi Wang
- Center for Microscopy and Analysis, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Xiaolin Wang
- Macau Institute of Materials Science and Engineering, Faculty of Innovation Engineering, School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, 999078, China
| | - Hao Wu
- Macau Institute of Materials Science and Engineering, Faculty of Innovation Engineering, School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, 999078, China
| | - Shuang Li
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Sichuan University, Chengdu, 610065, China
| | - Chong Cheng
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Sichuan University, Chengdu, 610065, China
- Department of Endodontics, State Key Laboratory of Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Institute of Chemistry and Biochemistry, Free University of Berlin, 14195, Berlin, Germany
| | - Changsheng Zhao
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
10
|
Wang Z, Li Z, Yang R, Wang Y, Ma W, Lv X, Gao J, Li Y. Rubidium ions as a novel therapeutic approach for glioblastoma. Sci Rep 2025; 15:12917. [PMID: 40234687 PMCID: PMC12000443 DOI: 10.1038/s41598-025-97688-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
Glioblastoma (GBM) is a highly aggressive brain tumor with limited treatment options, mainly due to challenges such as incomplete resection and blood-brain barrier limitations. Rubidium, a naturally occurring alkali metal with favorable biocompatibility, widely used in myocardial and tumor perfusion imaging as a blood flow tracer, is repurposed in this study to investigate its potential therapeutic effects and mechanisms against GBM. The impacts of rubidium ions (Rb⁺) on GBM cells were assessed through functional assays evaluating proliferation, migration, invasion, and apoptosis. RNA sequencing and Western blot analyses were employed to investigate molecular mechanisms, while in vivo models were used to evaluate therapeutic efficacy and safety. Rb⁺ treatment significantly suppressed GBM cell proliferation, migration, and invasion, while inducing apoptosis and cell cycle arrest at the G2/M phase. Mechanistic studies revealed that Rb⁺ downregulated the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway, contributing to the induction of apoptosis in tumor cells. In vivo, Rb⁺ exhibited potent anti-tumor activity with no detectable adverse effects on major organs, physiological functions, or behavior in mice. Our findings highlight Rb⁺ as a promising and innovative candidate for GBM therapy, leveraging the PI3K/AKT/mTOR pathway to inhibit tumor growth and promote apoptosis. This study underscores the potential of Rb⁺ in addressing the urgent need for novel GBM treatments, warranting further preclinical and clinical investigations.
Collapse
Affiliation(s)
- Zairan Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China
| | - Zhimin Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China
| | - Ran Yang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China
| | - Xiang Lv
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Jun Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China.
| | - Yongning Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China.
| |
Collapse
|
11
|
Guo C, Lin L, Wang Y, Jing J, Gong Q, Luo K. Nano drug delivery systems for advanced immune checkpoint blockade therapy. Theranostics 2025; 15:5440-5480. [PMID: 40303342 PMCID: PMC12036873 DOI: 10.7150/thno.112475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been widely utilized in the first-line therapy of various types of cancer. However, immune-related adverse events (irAEs) and resistance to ICIs remain intractable challenges for immune checkpoint blockade (ICB) therapy during clinic treatment. Nano drug delivery systems (NDDSs) have shown promising potential to improve anticancer efficacy and reduce side effects of small molecular drugs. The combination of nanotechnology and ICB provides new opportunities to overcome the challenges of immunotherapy. Nanoplatforms have been employed for direct delivery of ICIs, and they are preferred vehicles for combination therapy of ICIs and other therapeutic agents. In this review, the strategies of using NDDSs for advancing ICB therapy in recent years are surveyed, emphasizing the employment of NDDSs for combination treatment by ICIs and other agents to manipulate antitumor immunity. Analysis of current strategies for applying NDDSs for ICB leads to future research directions and development trends.
Collapse
Affiliation(s)
- Chenqi Guo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Lin
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yihan Wang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- West China School of Medicine, Chengdu 610041, China
| | - Jing Jing
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Xiamen Key Lab of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
12
|
Guan X, Shen Y, Zhao C, Li X, Li X, Lu D, Wang L, Liu L, Wu S, Huang B, Guo L, Xu H. Cascade-Responsive Nanoprodrug Disrupts Immune-Fibroblast Communications for Potentiated Cancer Mechanoimmunotherapy. Adv Healthc Mater 2025; 14:e2500176. [PMID: 40079115 DOI: 10.1002/adhm.202500176] [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: 01/10/2025] [Revised: 03/02/2025] [Indexed: 03/14/2025]
Abstract
The abnormal tumor mechanical microenvironment due to specific cancer-associated fibroblasts (CAFs) subset and low tumor immunogenicity caused by inefficient conversion of active chemotherapeutic agents are two key obstacles that impede patients with desmoplastic tumors from achieving stable and complete immune responses. Herein, it is demonstrated that FAP-α+CAFs-induced stromal stiffness accelerated tumor progression by precluding cytotoxic T lymphocytes. Subsequently, a cascade-responsive nanoprodrug capable of re-educating FAP-α+CAFs and amplifying tumor immunogenicity for potentiated cancer mechanoimmunotherapy is ingeniously designed. Benefiting from the active targeted release of angiotensin II receptor antagonist (losartan) guided by FAP-α cleavable peptide and the efficient conversion of topoisomerase I inhibitor (7-Ethyl-10-hydroxycamptothecin) prodrug under high glutathione/esterase within tumor cells, this regimen created an immune-activated landscape that retarded primary tumor growth and counteracted resistance to immune checkpoint inhibitor in mice with triple-negative breast cancer. This nanoprodrug-assisted mechanoimmunotherapy can serve as a universal strategy for conferring efficient tumoricidal immunity in "immune excluded" desmoplastic tumor interventions.
Collapse
Affiliation(s)
- Xin Guan
- Department of Ultrasound, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361000, P. R. China
| | - Yuting Shen
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Chongke Zhao
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Xiao Li
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Xiaolong Li
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Dan Lu
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Lifan Wang
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Linna Liu
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Shengbo Wu
- Department of Ultrasound, Zhejiang Hospital, Hangzhou, 310013, P. R. China
| | - Bin Huang
- Department of Ultrasound, Zhejiang Hospital, Hangzhou, 310013, P. R. China
| | - Lehang Guo
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Huixiong Xu
- Department of Ultrasound, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361000, P. R. China
| |
Collapse
|
13
|
Zhang R, Chen M, Zhou H, Liu Y, Wang Y, Chen C, Li Y, Zeng J, Cui J, Duan R, Gao M. Eliminating Radioresistance With a Magnetic Ion-Generator by Simultaneously Augmenting DNA Damage and Diminishing Immunosuppression. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2406378. [PMID: 39996275 DOI: 10.1002/adma.202406378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 01/23/2025] [Indexed: 02/26/2025]
Abstract
Radiotherapy (RT) hinges on DNA damage-induced cancer cell death and the subsequent anti-tumor immunity. However, the efficacy of RT is curtailed by cell cycle heterogeneity and an immunosuppressive tumor microenvironment, which foster radioresistance. Here an ion generator-based RT enhancement strategy is demonstrated in a mouse model of the radioresistant tumor. The ion generator is degraded in the tumor microenvironment, resulting in iron-triggered ferroptosis that enhanced immunogenic cell death and a manganese-activated stimulator of interferon gene that reversed the immunosuppressive environment. As a result, the proposed strategy promotes dendritic cells maturity, augmentes CD8+ T cell infiltration of tumors, suppresses intratumoral myeloid-derived suppressor cells, and limits the M2 macrophages polarization, indicating the formation of an immunoreactive microenvironment. Significantly, this approach impedes the growth of not just primary, but also distal metastatic tumors. It is thus believed that the current ion generator provides a robust and enduring countermeasure to radioresistant cancer and its metastasis, with potential implications for enhancing the efficacy of RT in clinically resistant tumors.
Collapse
Affiliation(s)
- Ruru Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Mei Chen
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Hui Zhou
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Yan Liu
- The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yi Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Can Chen
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Yueping Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Jiabin Cui
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Ruixue Duan
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Mingyuan Gao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
- The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| |
Collapse
|
14
|
Xue H, Jin J, Huang X, Tan Z, Zeng Y, Lu G, Hu X, Chen K, Su Y, Hu X, Peng X, Jiang L, Wu J. Wearable flexible ultrasound microneedle patch for cancer immunotherapy. Nat Commun 2025; 16:2650. [PMID: 40102412 PMCID: PMC11920228 DOI: 10.1038/s41467-025-58075-z] [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: 08/21/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
Clinical approaches for cancer therapy face several interrelated challenges involving inefficient drug delivery, potential adverse side effects, and inconvenience. Here, we present an integrated wearable flexible ultrasound microneedle patch (wf-UMP) that serves as a portable platform for convenient, efficient, and minimally invasive cancer therapy. The wf-UMP adopts an all-in-one bioelectronic concept, which integrates a stretchable lead-free ultrasound transducer array for acoustic emission, a bioadhesive hydrogel elastomer for robust adhesion and acoustic coupling, and a dissolvable microneedle patch loaded with biocompatible piezoelectric nanoparticles for painless drug delivery and reactive oxygen species generation. With soft mechanical properties and enhanced electromechanical performance, wf-UMP can be robustly worn on curved and dynamic tissue surfaces for easy and effective manipulation. In preclinical studies involving mice, wf-UMP demonstrated notable anticancer effects by inducing tumor cell apoptosis, amplifying oxidative stress, and modulating immune cell proliferation. Furthermore, the synergistic immunotherapy induced by wf-UMP and Anti-PD1 further improved anticancer immunity by activating immunogenic cell death and regulating macrophages polarization, inhibiting distant tumor growth and tumor recurrence.
Collapse
Affiliation(s)
- Haoyue Xue
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610064, China
| | - Jing Jin
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xing Huang
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610064, China
| | - Zhi Tan
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610064, China
| | - Yushun Zeng
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Gengxi Lu
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Xin Hu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Keliang Chen
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yonglin Su
- Department of Rehabilitation, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaolin Hu
- West China School of Nursing, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Laiming Jiang
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610064, China.
| | - Jiagang Wu
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610064, China.
- College of Physics, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
15
|
Meng Z, Li J, Wang H, Cao Z, Lu W, Niu X, Yang Y, Li Z, Wang Y, Lu S. NLRP4 unlocks an NK/macrophages-centered ecosystem to suppress non-small cell lung cancer. Biomark Res 2025; 13:44. [PMID: 40087771 PMCID: PMC11909883 DOI: 10.1186/s40364-025-00756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Tumor immune evasion extends beyond T cells, affecting innate immune elements like natural killer cells (NK) and macrophages within the tumor-immune microenvironment (TIME). Nevertheless, translational strategies to trigger collaboration of NK cells and macrophages to initiate sufficient anti-tumor cytoxicity remain scarce and are urgently needed. METHODS In this study, TCGA datasets was used to confirm the prognosis value of the expression level of NLR family pyrin domain containing 4 (NLRP4) in NSCLC and the tumor tissues microarray was used to further check its clinical-relevance at protein-level. Subsequently, a tumor cell line with stable NLRP4 overexpression was established and subcutaneous tumor models in C57BL/6J mice were used to validate the anti-tumor characteristics of NLRP4. After analyzing the tumor microenvironment using flow cytometry and multiplex immunofluorescence, we further validated our findings through co-culture transwell assays and TCGA analysis. Utilizing bulk-RNA sequencing, proteomics, and mass spectrometry of mouse tumor tissues, we innovatively identified the downstream pathways of NLRP4 and verified them through co-immunoprecipitation (co-IP) and Western blot (WB) experiments. RESULTS NLRP4 could trigger a distinct anti-tumor ecosystem organized by TIGIT+TNFA+ NK and iNOS+ M1 in lung cancer, discovered in TCGA analysis and verified in murine model. NLRP4-eco exerted tumor-suppression capacity through chemokine reprogramming including CCL5 and CXCL2. Meanwhile, the cytoxicity of NK could be facilitated by iNOS+M1. Mechanistically, NLRP4 stimulated PI3K/Akt-NF-kB axis through suppression of the activity of PP2A. Besides, knockdown of CCL5 and blockade of CXCL2-CXCR2 axis abolished chemotaxis of TIGIT+TNFA+ NK and iNOS+ M1 respectively, as well as for LB-100, a PP2A inhibitor. CONCLUSION Altogether, we delineated NLRP4's unexplored facets and discovered an NLRP4-driven anti-tumor ecosystem composed of TIGIT+TNFA+ NK and iNOS+ M1. Finally, targeting PP2A by its inhibitor successfully mimicked the anti-tumor capacity of the overexpression of NLRP4.
Collapse
Affiliation(s)
- Zhouwenli Meng
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Jian Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Hui Wang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Zhengqi Cao
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Wenqing Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Xiaomin Niu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Yi Yang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
| | - Ying Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China.
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
| |
Collapse
|
16
|
Sun Y, Liu L, He H, Cui G, Zheng Y, Ye C, Qu L, Sun Y, Ji J, Lammers T, Zhang Y, Zhong Z. Co-activating STING-TLR9 pathways promotes radiotherapy-induced cancer vaccination. J Control Release 2025; 379:327-343. [PMID: 39778743 DOI: 10.1016/j.jconrel.2024.12.079] [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: 11/08/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Vaccination may cure cancer patients by inducing tumor-specific immune responses. Radiotherapy is an appealing strategy to generate cancer vaccines in situ; thus far, however, only modest and short-lived immune responses are achieved. We here show that radiation combined with co-activating STING-TLR9 can generate powerful in situ cancer vaccines. Notably, radiation at a dose of 12Gy is found to be optimal for boosting tumor cell immunogenicity, and STING-TLR9 co-stimulation by a dual immune activation nano-agonist overrides key immunosuppressive effects associated with radiotherapy. Local radiotherapy combined with the dual immune activation nano-agonists elicits strong systemic anti-tumor immune responses, resulting in complete regression of tumors and metastases in multiple syngeneic murine tumor models. This work introduces a novel and highly potent cancer immunotherapeutic strategy that holds promise for the personalized treatment of intractable cancers.
Collapse
Affiliation(s)
- Yu Sun
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Liang Liu
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; School of Optical and Electronic Information, Suzhou City University, Suzhou 215104, People's Republic of China
| | - Huilan He
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Guanhong Cui
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Yun Zheng
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Chunlian Ye
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Liping Qu
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Yinping Sun
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Jinlong Ji
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Clinic, Aachen 52074, Germany
| | - Ying Zhang
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China.
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
17
|
Xing Y, Li J, Wang L, Zhu Z, Yan J, Liu Y, Liu Q. A Bifunctional Lysosome-Targeting Chimera Nanoplatform for Tumor-Selective Protein Degradation and Enhanced Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417942. [PMID: 39888098 DOI: 10.1002/adma.202417942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/09/2025] [Indexed: 02/01/2025]
Abstract
Lysosome-targeting chimeras (LYTACs) have recently emerged as a promising therapeutic strategy for degrading extracellular and membrane-associated pathogenic proteins by hijacking lysosome-targeting receptors. However, the antitumor performance of LYTAC is limited by its insufficient tumor accumulation and nonspecific activation. Additionally, the synergistic effects of LYTACs and other therapeutic modalities are crucial. To address these issues, a bifunctional LYTAC nanoplatform (NLTC) is developed for tumor-selective protein degradation and enhanced cancer immunotherapy. By rationally controlling the surface composition, the NLTC can effectively transport extracellular or membrane proteins into lysosomes for degradation via cation-independent mannose 6-phosphate receptors. With removable surface modification, an NLTC is obtained that efficiently accumulated in tumor tissues and avoided on-target off-tumor toxicity. Moreover, the synthesis method of NLTC is generally applicable to various enzymes. Thus, catalase (CAT) is encapsulated with NLTC to synergistically degrade cancer cell surface programmed death ligand-1 (PD-L1), relieve the immunosuppressive tumor microenvironment for effective cancer immunotherapy, and significantly inhibit tumor growth, recurrence, and metastasis in B16F10-bearing mice. This work presents a bifunctional LYTAC nanoplatform that can not only perform tissue-selective protein degradation but also integrate other therapeutic modalities, providing insights into the design of advanced LYTAC technologies for clinical applications.
Collapse
Affiliation(s)
- Yumeng Xing
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
- School of Pharmacy, Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Jingjing Li
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Leyuan Wang
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
- School of Pharmacy, Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Zhihui Zhu
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
- School of Pharmacy, Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Jian Yan
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
- School of Pharmacy, Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Yang Liu
- College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Qi Liu
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
- School of Pharmacy, Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
18
|
Jia M, Lu R, Li P, Liao X, Tan Y, Zhang S. Inflammation-reducing thermosensitive hydrogel with photothermal conversion for skin cancer therapy. J Control Release 2025; 378:377-389. [PMID: 39701451 DOI: 10.1016/j.jconrel.2024.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Photothermal therapy (PTT) has widely been utilized for postoperative treatment of skin cancer, while high temperature, usually >50 °C, would induce damage to healthy tissue and increased wound inflammation. Herein, we developed an "all in one" hydrogel to enhance mild PTT for postoperative skin cancer treatment while circumventing photothermo-induced inflammation by loading quercetin (Que)-coated tannin‑iron (TA-Fe) nanoparticles with poly (N-acrylylglycine) amine (PNAGA) hydrogel (Que@TA-Fe@PNAGA). Exposure to near-infrared light, Que.@TA-Fe@PNAGA occurred a mild temperature increase (∼47 °C), which induces local mild PTT and disrupts the hydrogen bonds within the hydrogel, triggering a gel-to-sol phase transition and the release of Que.@TA-Fe nanoparticles. These released nanoparticles inhibit the expression of heat shock proteins in tumor cells by producing reactive oxygen species and enter inflammatory cells to release TA and Que. via acid hydrolysis, reducing tumor necrosis factor-α expression by 66.6 % and promoting M1-to-M2 macrophage conversion. Based on this integrated functionality, Que.@TA-Fe@PNAGA hydrogel achieves over 99.4 % tumor inhibition rate, effectively avoids photothermo-induced damage in normal tissue and inflammation, and thus represents a new approach for postoperative photothermal therapy in skin cancer treatment.
Collapse
Affiliation(s)
- Mengqi Jia
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; School of Basic Medical Science, Henan University, Zhengzhou 450046, China
| | - Ruilin Lu
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Pengfei Li
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xiaoming Liao
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | - Yanfei Tan
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Shiyong Zhang
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
19
|
He M, Chen S, Yu H, Fan X, Wu H, Wang Y, Wang H, Yin X. Advances in nanoparticle-based radiotherapy for cancer treatment. iScience 2025; 28:111602. [PMID: 39834854 PMCID: PMC11743923 DOI: 10.1016/j.isci.2024.111602] [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] [Indexed: 01/22/2025] Open
Abstract
Radiotherapy has long been recognized as an effective conventional approach in both clinical and scientific research, primarily through mechanisms involving DNA destruction or the generation of reactive oxygen species to target tumors. However, significant challenges persist, including the unavoidable damage to normal tissues and the development of radiation resistance. As a result, nanotechnology-based radiotherapy has garnered considerable attention for its potential to enhance precision in irradiation, improve radiosensitization, and achieve therapeutic advancements. Importantly, radiotherapy alone frequently falls short of fully eradicating tumors. Consequently, to augment the efficacy of radiotherapy, it is often integrated with other therapeutic strategies. This review elucidates the mechanisms of radiotherapy sensitization based on diverse nanoparticles. Typically, radiotherapy is sensitized through augmenting reactive oxygen species production, targeted radiotherapy, hypoxia relief, enhancement of antitumor immune microenvironment, and G2/M cell cycle arrest. Moreover, the incorporation of nanoparticle-based anti-tumor strategies with radiotherapy markedly enhances the current state of radiotherapy. Additionally, a compilation of clinical trials utilizing nano-radioenhancers is presented. Finally, future prospects for clinical translation in this field are thoroughly examined.
Collapse
Affiliation(s)
- Meijuan He
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Shixiong Chen
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
| | - Hongwei Yu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xuhui Fan
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Hong Wu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yihui Wang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
| | - Han Wang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
- Jiading Branch of Shanghai General Hospital, Shanghai 201803, China
| | - Xiaorui Yin
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
20
|
Guo Z, Zheng H, Wang T, Han N, Zhang H, Li J, Cheng X, Ye J, Du S, Li P. Combination Nanodrug Delivery Systems Facilitate the Syncretism of Chemotherapy with Immunotherapy to Promote Cancer Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405752. [PMID: 39544164 DOI: 10.1002/smll.202405752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/12/2024] [Indexed: 11/17/2024]
Abstract
Cancer has emerged as a significant threat that gravely endanger human health. Anti-tumor immunotherapy has now emerged as an important treatment for cancer. However, immunosuppressive tumor microenvironment limits the antitumor immunity. The importance of the immune system in the cancer treatment process must be emphasized. Herein, two precision-targeted nanoparticles PD-L1@Cur-NPs and PD-1@AS-NPs are constructed for cancer treatment. PD-L1@Cur-NPs can precisely target tumor cells in vivo to eradicate tumor cells or induce them apoptosis. PD-1@AS-NPs can precisely target T cells in vivo to activate the T cell-mediated immune system and induce antitumor immune responses. Furthermore, these two nanoparticles have good synergistic effect and show stronger antitumor effect after combination. After treatment with the combination of two nanoparticles, the tumor volumes of C57BL/6 tumor-bearing mice are significantly reduced. Moreover, the percentage of CD8+T cells and CD4+T cells in the tumor significantly increased, and the percentage of regulatory T cells significantly decreased. The percentage of memory T cells and memory effector T cells in the spleen also significantly increased after treatment, suggesting that the antitumor immunity is activated after treatment. This study provides a new antitumor treatment strategy combining chemotherapy and immunotherapy, which has good application prospect.
Collapse
Affiliation(s)
- Zishuo Guo
- Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Haocheng Zheng
- Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Tieshan Wang
- Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Ning Han
- Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Haitong Zhang
- Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jialing Li
- Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xuehao Cheng
- Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jinhong Ye
- Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Shouying Du
- Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Pengyue Li
- Beijing University of Chinese Medicine, Beijing, 102488, China
| |
Collapse
|
21
|
Wang LJ, Zhao Y, Wang YS, Xing L, Duan YJ, Zang HH. The IL-10/STAT3 Axis Nasopharyngeal Carcinoma Cancer stem cell and radio resistance. Sci Rep 2024; 14:31943. [PMID: 39738457 PMCID: PMC11685382 DOI: 10.1038/s41598-024-83423-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
One of the primary reasons for the failure of therapy in nasopharyngeal cancer (NPC) is radio resistance-related localized one, which may lead to tumor residuals or recurrences. Several studies have linked interleukin-10 (IL-10) to crucial functions in cancer development and response to therapy. Its function in NPC's radio resistance is, however, not well understood. Enzyme-linked immunosorbent assay (ELISA) and quantitative real-time PCR were utilized for confirming IL-10 expression in NPC cell lines. The prognostic significance of IL-10 was also assessed via Kaplan-Meier analysis. CNE2R, a radioresistant NPC cell line, expressed IL-10 at high levels, which were also shown to be considerably elevated in individuals with radioresistant NPC, as measured by ELISA. Moreover, the levels were also linked to poor clinical outcomes and prognosis in cancer cases. We also showed some evidence of a link between hypoxia-inducible factor 1-alpha (HIF-1 A) and serum IL-10 levels in NPC. Meanwhile, we find that IL-10 is up-regulated in CSC. IL-10 enhanced the self-renewal and tumorigenesis of nasopharyngeal CSC. In terms of mechanism, IL-10 enhances nasopharyngeal CSC self-renewal and tumorigenesis by activating STAT3 pathway. In NPC, IL-10/STAT3 Axis Nasopharyngeal Carcinoma Cancer stem cell and radio resistance.
Collapse
Affiliation(s)
- Li-Jun Wang
- Department of Otolaryngology Head and Neck Surgery, Shijiazhuang People's Hospital, Shijiazhuang, Hebei Province, China
| | - Ying Zhao
- Department of Otolaryngology Head and Neck Surgery, Shijiazhuang People's Hospital, Shijiazhuang, Hebei Province, China
| | - Yan-Sheng Wang
- Department of Nuclear Medicine, General Hospital of Shenzhen University, Shenzhen, Guangdong Province, China
| | - Lei Xing
- Department of Otolaryngology Head and Neck Surgery, Shijiazhuang People's Hospital, Shijiazhuang, Hebei Province, China
| | - Yun-Jing Duan
- Department of Otolaryngology Head and Neck Surgery, Shijiazhuang People's Hospital, Shijiazhuang, Hebei Province, China
| | - Huan-Huan Zang
- Department of Otolaryngology Head and Neck Surgery, Shijiazhuang People's Hospital, Shijiazhuang, Hebei Province, China.
| |
Collapse
|
22
|
Hong S, Park J, Oh Y, Cho H, Kim K. Nanotechnology-Based Strategies for Safe and Effective Immunotherapy. Molecules 2024; 29:5855. [PMID: 39769944 PMCID: PMC11676242 DOI: 10.3390/molecules29245855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer immunotherapy using immune checkpoint blockades has emerged as a promising therapeutic approach. However, immunotherapy faces challenges such as low response rates in solid tumors, necessitating strategies to remodel the immune-suppressive tumor microenvironment (TME) into an immune-activated state. One of the primary approaches to achieve this transformation is through the induction of immunogenic cell death (ICD). Herein, we discussed strategies to maximize ICD induction using nanoparticles. In particular, this review highlighted various studies integrating chemotherapy, radiation therapy (RT), photodynamic therapy (PDT), and photothermal therapy (PTT) with nanoparticle-based immunotherapy. The research covered in this review aims to provide valuable insights for future studies on nanoparticle-assisted immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; (S.H.); (J.P.); (Y.O.); (H.C.)
| |
Collapse
|
23
|
Hiep Tran T, Thu Phuong Tran T. Current status of nanoparticle-mediated immunogenic cell death in cancer immunotherapy. Int Immunopharmacol 2024; 142:113085. [PMID: 39276455 DOI: 10.1016/j.intimp.2024.113085] [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: 06/24/2024] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024]
Abstract
Immunogenic cell death (ICD) encompasses various forms of cell death modalities, including apoptosis, necroptosis, ferroptosis, and pyroptosis. It arises from a harmonious interplay of adjuvant (damage-associated molecular patterns-DAMPs and chemokines/cytokines) and antigenicity (tumor-associated antigens-TAA) to induce immune-reaction toward cancer cells. Inducing ICD stands out as a promising approach in cancer immunotherapy, capable of directly eliminating cancer cells and of eliciting enduring antitumor immune responses. Conventional tumor therapies like radiation therapy, photodynamic therapy, and chemotherapy can also induce ICD which could amplify their activities. The development of effective ICD inducers like nano-systems is crucial for ensuring safe and efficacious immunotherapy. Nanoparticles hold considerable promise in cancer therapy, offering enhanced therapeutic outcomes and mitigated side effects. They could be the capacity to adjust systemic biodistribution, augment the accumulation of therapeutic agents at the intended site and protect active agents from the complexity of human biofluid. This review aims to outline the role of nanoparticles in triggering ICD for cancer immunotherapy that potentially pave the way for cancer treatment.
Collapse
Affiliation(s)
- Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam
| | - Thi Thu Phuong Tran
- Department of Life Sciences, University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam.
| |
Collapse
|
24
|
Wang Z, Han X, Sun G, Yu M, Qin J, Zhang Y, Ding D. Advances in cancer diagnosis and therapy by alginate-based multifunctional hydrogels: A review. Int J Biol Macromol 2024; 283:137707. [PMID: 39566758 DOI: 10.1016/j.ijbiomac.2024.137707] [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: 10/11/2024] [Revised: 10/30/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
The field of oncology has been changed by the application of hydrogels. These 3D polymeric networks have demonstrated significant promise in the treatment of cancer and can boost the efficacy of conventional therapeutics including chemotherapy and immunotherapy. Noteworthy, the development of biocompatible and effective hydrogels has been of interest. In this case, alginate as a biopolymer and carbohydrate polymer has been used to modify or synthesis multifunctional nanoparticles for the treatment of human diseases, especially cancer. Therefore, highlighting the function of alginate in the development of hydrogels in cancer therapy can provide new insights for improving outcome and survival rate of patients. Alginate hydrogels improve the specific and selective delivery of cargo and therefore, they reduce the systemic toxicity of drugs, while they enhance anti-cancer activity. Alginate hydrogels protect the genes against degradation by enzymes and increase blood circulation time. The alginate hydrogels can respond to the specific stimuli in the tumor microenvironment including pH, redox and light to improve the site-specific release of cargo. The nanoparticles can be incorporated in the structure of alginate hydrogels to augment their anti-cancer activity. In addition, alginate hydrogels can accelerate immunotherapy and phototherapy through delivery of immunomodulators and photosensitizers, respectively.
Collapse
Affiliation(s)
- Ziwen Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xu Han
- Department of Emergency, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Guowei Sun
- Interventional Center, Fengcheng Central Hospital, Fengcheng 118199, China
| | - Miao Yu
- Department of Respiratory, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Juan Qin
- Department of Endocrinology and Metabolism, Shenyang Fourth People Hospital, Shenyang 110001, China
| | - Yuting Zhang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Ding Ding
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
25
|
Qi L, Li Z, Liu J, Chen X. Omics-Enhanced Nanomedicine for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409102. [PMID: 39473316 DOI: 10.1002/adma.202409102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/10/2024] [Indexed: 12/13/2024]
Abstract
Cancer nanomedicine has emerged as a promising approach to overcome the limitations of conventional cancer therapies, offering enhanced efficacy and safety in cancer management. However, the inherent heterogeneity of tumors presents increasing challenges for the application of cancer nanomedicine in both diagnosis and treatment. This heterogeneity necessitates the integration of advanced and high-throughput analytical techniques to tailor nanomedicine strategies to individual tumor profiles. Omics technologies, encompassing genomics, epigenomics, transcriptomics, proteomics, metabolomics, and more, provide unparalleled insights into the molecular and cellular mechanisms underlying cancer. By dissecting tumor heterogeneity across multiple levels, these technologies offer robust support for the development of personalized and precise cancer nanomedicine strategies. In this review, the principles, techniques, and applications of key omics technologies are summarized. Especially, the synergistic integration of omics and nanomedicine in cancer therapy is explored, focusing on enhanced diagnostic accuracy, optimized therapeutic strategies and the assessment of nanomedicine-mediated biological responses. Moreover, this review addresses current challenges and outlines future directions in the field of omics-enhanced nanomedicine. By offering valuable insights and guidance, this review aims to advance the integration of omics with nanomedicine, ultimately driving improved diagnostic and therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
| | - Jianping Liu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xiaoyuan Chen
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore
| |
Collapse
|
26
|
Duan X, Xue B, Xu Z, Niu Z. Multimodal Photodynamic Therapy by Inhibiting the Nrf2/ARE Signaling Pathway in Tumors. ACS Biomater Sci Eng 2024; 10:7018-7029. [PMID: 39417585 DOI: 10.1021/acsbiomaterials.4c01643] [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: 10/19/2024]
Abstract
Photodynamic therapy (PDT) has been widely used in the clinical therapy of various tumors, especially superficial tumors. However, the tumor microenvironment presents hypoxia, as well as the inherent antioxidant system (e.g., Nrf2) of tumor cells limits the therapeutic outcomes. Herein, a cascade-responsive "oxidative stress amplifier" (named EZ@TD) is designed by encapsulating manganese-doped carbon dots acting as a photosensitizer and catalase (CAT)-like nanozyme within pH-sensitive ZIF-8 and Zn2+-activated DNAzyme for relieving hypoxia and efficient Nrf2 gene disruption to enhance PDT. It is demonstrated that EZ@TD synergistically inhibited tumor growth and activated the antitumor immune response by inhibiting the Nrf2/ARE signaling pathway in tumors. We provide a new paradigm for amplifying intracellular oxidative stress by interfering with various signaling pathways.
Collapse
Affiliation(s)
- Xin Duan
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Bingjian Xue
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zimeng Xu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zixu Niu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
27
|
Hou X, Shen Y, Huang B, Li Q, Li S, Jiang T, Shan X, Xu W, Liu S, Wu S, Zhao D, Zhu A, Sun L, Xu H, Yue W. Losartan-based nanocomposite hydrogel overcomes chemo-immunotherapy resistance by remodeling tumor mechanical microenvironment. J Nanobiotechnology 2024; 22:667. [PMID: 39472933 PMCID: PMC11523888 DOI: 10.1186/s12951-024-02871-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Preclinical studies demonstrating high cure rates with PD1/PD-L1 combinations have led to numerous clinical trials, but emerging results are disappointing. These combined immunotherapies are commonly employed for patients with refractory tumors following prior treatment with cytotoxic agents. Here, we uncovered that the post-chemotherapy tumor presents a unique mechanical microenvironment characterized by an altered extracellular matrix (ECM) elasticity and increased stiffness, which facilitate the development of aggressive tumor phenotypes and confer resistance to checkpoint blocking therapy. As thus, we rationally designed an in situ nanocomposite hydrogel system, LOS&FeOX@Gel, which enabled effective and specific delivery of the therapeutic payloads (losartan [LOS] and oxaliplatin [OX]) into tumor. We demonstrate that sustained release of LOS effectively remodels the tumor mechanical microenvironment (TMM) by reducing ECM deposition and its associated "solid stress", thereby augmenting the efficacy of OX and its immunological effects. Importantly, this hydrogel system greatly sensitized post-chemotherapy tumor to checkpoint blocking therapy, showing synergistic therapeutic effects against cancer metastasis. Our study provides mechanistic insights and preclinical rationale for modulating TMM as a potential neoadjuvant regimen for tumor to optimize the benefits of chemo-immunotherapy, which lays the groundwork for leveraging "mechanical-immunoengineering" strategies to combat refractory tumors.
Collapse
Affiliation(s)
- Xiaodong Hou
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China
- Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, P.R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China
| | - Yuting Shen
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China
- Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, P.R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China
| | - Bin Huang
- Department of Ultrasound, Zhejiang Hospital, Hangzhou, Zhejiang Province, 310013, P.R. China
| | - Qiuyan Li
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China
- Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, P.R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China
| | - Shaoyue Li
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China
- Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, P.R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China
| | - Tingting Jiang
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China
- Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, P.R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China
| | - Xuexia Shan
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China
- Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, P.R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China
| | - Weichen Xu
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China
- Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, P.R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China
| | - Shuo Liu
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China
- Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, P.R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China
| | - Shengbo Wu
- Department of Ultrasound, Zhejiang Hospital, Hangzhou, Zhejiang Province, 310013, P.R. China
| | - De Zhao
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China
| | - Anqi Zhu
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China
- Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, P.R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China
| | - Liping Sun
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.
- Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, P.R. China.
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China.
| | - Huixiong Xu
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China.
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China.
| | - Wenwen Yue
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.
- Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, P.R. China.
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P.R. China.
| |
Collapse
|
28
|
Liu Z, Chen J, Ren Y, Liu S, Ba Y, Zuo A, Luo P, Cheng Q, Xu H, Han X. Multi-stage mechanisms of tumor metastasis and therapeutic strategies. Signal Transduct Target Ther 2024; 9:270. [PMID: 39389953 PMCID: PMC11467208 DOI: 10.1038/s41392-024-01955-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 07/18/2024] [Accepted: 08/24/2024] [Indexed: 10/12/2024] Open
Abstract
The cascade of metastasis in tumor cells, exhibiting organ-specific tendencies, may occur at numerous phases of the disease and progress under intense evolutionary pressures. Organ-specific metastasis relies on the formation of pre-metastatic niche (PMN), with diverse cell types and complex cell interactions contributing to this concept, adding a new dimension to the traditional metastasis cascade. Prior to metastatic dissemination, as orchestrators of PMN formation, primary tumor-derived extracellular vesicles prepare a fertile microenvironment for the settlement and colonization of circulating tumor cells at distant secondary sites, significantly impacting cancer progression and outcomes. Obviously, solely intervening in cancer metastatic sites passively after macrometastasis is often insufficient. Early prediction of metastasis and holistic, macro-level control represent the future directions in cancer therapy. This review emphasizes the dynamic and intricate systematic alterations that occur as cancer progresses, illustrates the immunological landscape of organ-specific PMN creation, and deepens understanding of treatment modalities pertinent to metastasis, thereby identifying some prognostic and predictive biomarkers favorable to early predict the occurrence of metastasis and design appropriate treatment combinations.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingqi Chen
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China.
| |
Collapse
|
29
|
Jiang Y, Wang C, Zu C, Rong X, Yu Q, Jiang J. Synergistic Potential of Nanomedicine in Prostate Cancer Immunotherapy: Breakthroughs and Prospects. Int J Nanomedicine 2024; 19:9459-9486. [PMID: 39371481 PMCID: PMC11456300 DOI: 10.2147/ijn.s466396] [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: 02/28/2024] [Accepted: 08/16/2024] [Indexed: 10/08/2024] Open
Abstract
Given the global prevalence of prostate cancer in men, it is crucial to explore more effective treatment strategies. Recently, immunotherapy has emerged as a promising cancer treatment due to its unique mechanism of action and potential long-term effectiveness. However, its limited efficacy in prostate cancer has prompted renewed interest in developing strategies to improve immunotherapy outcomes. Nanomedicine offers a novel perspective on cancer treatment with its unique size effects and surface properties. By employing targeted delivery, controlled release, and enhanced immunogenicity, nanoparticles can be synergized with nanomedicine platforms to amplify the effectiveness of immunotherapy in treating prostate cancer. Simultaneously, nanotechnology can address the limitations of immunotherapy and the challenges of immune escape and tumor microenvironment regulation. Additionally, the synergistic effects of combining nanomedicine with other therapies offer promising clinical outcomes. Innovative applications of nanomedicine include smart nanocarriers, stimulus-responsive systems, and precision medicine approaches to overcome translational obstacles in prostate cancer immunotherapy. This review highlights the transformative potential of nanomedicine in enhancing prostate cancer immunotherapy and emphasizes the need for interdisciplinary collaboration to drive research and clinical applications forward.
Collapse
Affiliation(s)
- Yueyao Jiang
- Department of Pharmacy, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Chengran Wang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Chuancheng Zu
- China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Xin’ao Rong
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Qian Yu
- Department of Pharmacy, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Jinlan Jiang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| |
Collapse
|
30
|
Li K, Li L, Xie X, Zhu J, Xia D, Xiang L, Cai K, Zhang J. Spatially confined photoacoustic effects of responsive nanoassembly boosts lysosomal membrane permeabilization and immunotherapy of triple-negative breast cancer. Acta Biomater 2024; 187:381-395. [PMID: 39209130 DOI: 10.1016/j.actbio.2024.08.021] [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: 06/13/2024] [Revised: 08/10/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Although immunogenic cell death (ICD) induced by lysosomal membrane permeabilization (LMP) evidently enhance the effectiveness of antitumor immunity for triple-negative breast cancer (TNBC) with poor immunogenicity, their potential is increasingly restricted by the development of other death pathways and the repair of lysosomes by endoplasmic reticulum (ER) during LMP induction. Herein, a polydopamine nanocomposite with i-motif DNA modified and BNN6 loaded is prepared toward boosting LMP and immunotherapy of TNBC by synergy of spatially confined photoacoustic (PA) effects and nitric oxide. Combining the high-frequency pulsed laser (4000 kHz) with the intra-lysosomal assembly of nanocomposites produced spatially confined and significantly boosted PA effects (4.8-fold higher than the individually dispersed particles extracellular), suppressing damage to other cellular components and selectively reducing lysosomal integrity to 19.2 %. Simultaneously, the releasing of nitric oxide inhibited the repair of lysosomes by ER stress, causing exacerbated LMP. Consequently, efficient immune activation was achieved, including the abundant releasing of CRT/HMGB1 (5.93-6.8-fold), the increasing maturation of dendritic cells (3.41-fold), and the fostered recruitment of CD4+/CD8+T cells (3.99-3.78-fold) in vivo. The study paves a new avenue for the rational design and synergy of confined energy conversion and responsive nanostructures to achieve the treatment of low immunogenicity tumors. STATEMENT OF SIGNIFICANCE: A strategy of boosting lysosomal membrane permeabilization (LMP) and concomitantly preventing the repair was developed to address the immunotherapy challenge of triple-negative breast cancer. Spatially confined and significantly enhanced photoacoustic (PA) effects were achieved through DNA-guided pH-responsive assembly of polydopamine nanocomposites in lysosomes and application of a high-frequency pulsed laser. Efficient immunogenic cell death was guaranteed by selective and powerful damage of lysosomal membranes through the significant contrast of PA intensities for dispersed/assembled particles and nitric oxide release induced endoplasmic reticulum stress. The study paves a new avenue for the rational design and synergy of confined energy conversion and responsive nanostructures to achieve the treatment of low immunogenicity tumors.
Collapse
Affiliation(s)
- Kunlin Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No 174 Shazheng Road, Chongqing 400044, China
| | - Lin Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No 174 Shazheng Road, Chongqing 400044, China
| | - Xiyue Xie
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No 174 Shazheng Road, Chongqing 400044, China
| | - Jing Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No 174 Shazheng Road, Chongqing 400044, China
| | - Daqing Xia
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No 174 Shazheng Road, Chongqing 400044, China
| | - Lunli Xiang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No 174 Shazheng Road, Chongqing 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No 174 Shazheng Road, Chongqing 400044, China
| | - Jixi Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No 174 Shazheng Road, Chongqing 400044, China.
| |
Collapse
|
31
|
Di Y, Yang Z, Song G, Shen Q, Bai H, Huang Y, Lv F, Wang S. Biosynthesis of multifunctional transformable peptides for downregulation of PD-L1. Chem Commun (Camb) 2024; 60:10938-10941. [PMID: 39258452 DOI: 10.1039/d4cc03146f] [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: 09/12/2024]
Abstract
Here, we present a biosynthesized material M1 for immune checkpoint blocking therapy. M1 could realize a morphological transformation from globular to fibrous in situ in the presence of cathepsin B (CtsB) after entering tumor cells. The GO203 peptides of M1 are exposed, which could bind to mucin 1 (MUC1) to suppress the homodimerization process of MUC1, thereby downregulating PD-L1 expression.
Collapse
Affiliation(s)
- Yufei Di
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiwen Yang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gang Song
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Shen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Haotian Bai
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Yiming Huang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Fengting Lv
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
32
|
Kim JH, Dareowolabi BO, Thiruvengadam R, Moon EY. Application of Nanotechnology and Phytochemicals in Anticancer Therapy. Pharmaceutics 2024; 16:1169. [PMID: 39339205 PMCID: PMC11435124 DOI: 10.3390/pharmaceutics16091169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/22/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer is well recognized as a leading cause of mortality. Although surgery tends to be the primary treatment option for many solid cancers, cancer surgery is still a risk factor for metastatic diseases and recurrence. For this reason, a variety of medications has been adopted for the postsurgical care of patients with cancer. However, conventional medicines have shown major challenges such as drug resistance, a high level of drug toxicity, and different drug responses, due to tumor heterogeneity. Nanotechnology-based therapeutic formulations could effectively overcome the challenges faced by conventional treatment methods. In particular, the combined use of nanomedicine with natural phytochemicals can enhance tumor targeting and increase the efficacy of anticancer agents with better solubility and bioavailability and reduced side effects. However, there is limited evidence in relation to the application of phytochemicals in cancer treatment, particularly focusing on nanotechnology. Therefore, in this review, first, we introduce the drug carriers used in advanced nanotechnology and their strengths and limitations. Second, we provide an update on well-studied nanotechnology-based anticancer therapies related to the carcinogenesis process, including signaling pathways related to transforming growth factor-β (TGF-β), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3 kinase (PI3K), Wnt, poly(ADP-ribose) polymerase (PARP), Notch, and Hedgehog (HH). Third, we introduce approved nanomedicines currently available for anticancer therapy. Fourth, we discuss the potential roles of natural phytochemicals as anticancer drugs. Fifth, we also discuss the synergistic effect of nanocarriers and phytochemicals in anticancer therapy.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea; (B.O.D.); (E.-Y.M.)
| | - Boluwatife Olamide Dareowolabi
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea; (B.O.D.); (E.-Y.M.)
| | - Rekha Thiruvengadam
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha Medical College, Saveetha University, Chennai 600077, India;
| | - Eun-Yi Moon
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea; (B.O.D.); (E.-Y.M.)
| |
Collapse
|
33
|
Liang X, Wang D, Zhao Y, Wang X, Yao S, Huang W, Yang Y, Dong X, Zhang L, Yang J. Tumor microenvironment-responsive manganese-based nano-modulator activate the cGAS-STING pathway to enhance innate immune system response. J Nanobiotechnology 2024; 22:535. [PMID: 39227944 PMCID: PMC11373498 DOI: 10.1186/s12951-024-02809-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Manganese ions (Mn2+) combined with adjuvants capable of damaging and lysing tumor cells form an antitumor nano-modulator that enhances the immune efficacy of cancer therapy through the cascade activation of the cyclic GMP-AMP interferon gene synthase-stimulator (cGAS-STING) pathway, which underscores the importance of developing antitumor nano-modulators, which induce DNA damage and augment cGAS-STING activity, as a critical future research direction. METHODS AND RESULTS: We have successfully synthesized an antitumor nano-modulator, which exhibits good dispersibility and biosafety. This nano-modulator is engineered by loading manganese dioxide nanosheets (M-NS) with zebularine (Zeb), known for its immunogenicity-enhancing effects, and conducting targeted surface modification using hyaluronic acid (HA). After systemic circulation to the tumor site, Mn2+, Zeb, and reactive oxygen species (ROS) are catalytically released in the tumor microenvironment by H+ and H2O2. These components can directly or indirectly damage the DNA or mitochondria of tumor cells, thereby inducing programmed cell death. Furthermore, they promote the accumulation of double-stranded DNA (dsDNA) in the cytoplasm, enhancing the activation of the cGAS-STING signalling pathway and boosting the production of type I interferon and the secretion of pro-inflammatory cytokines. Additionally, Zeb@MH-NS enhances the maturation of dendritic cells, the infiltration of cytotoxic T lymphocytes, and the recruitment of natural killer cells at the tumor site. CONCLUSIONS This HA-modified manganese-based hybrid nano-regulator can enhance antitumor therapy by boosting innate immune activity and may provide new directions for immunotherapy and clinical translation in cancer.
Collapse
Affiliation(s)
- Xiayi Liang
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Tumor Hospital of Guangxi Medical University, Guangxi Medical University, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Duo Wang
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 224001, China
| | - Yuanquan Zhao
- Department of Hepatobiliary, Pancreas and Spleen Surgery, the People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Xiaobo Wang
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Tumor Hospital of Guangxi Medical University, Guangxi Medical University, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Siyang Yao
- Department of Hepatobiliary, Pancreas and Spleen Surgery, the People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Wei Huang
- Department of Hepatobiliary, Pancreas and Spleen Surgery, the People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Yongyu Yang
- Department of Hepatobiliary, Pancreas and Spleen Surgery, the People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Xiaofeng Dong
- Department of Hepatobiliary, Pancreas and Spleen Surgery, the People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, China.
| | - Lei Zhang
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 224001, China.
| | - Jianrong Yang
- Department of Hepatobiliary, Pancreas and Spleen Surgery, the People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, China.
| |
Collapse
|
34
|
Su X, Li J, Xu X, Ye Y, Wang C, Pang G, Liu W, Liu A, Zhao C, Hao X. Strategies to enhance the therapeutic efficacy of anti-PD-1 antibody, anti-PD-L1 antibody and anti-CTLA-4 antibody in cancer therapy. J Transl Med 2024; 22:751. [PMID: 39123227 PMCID: PMC11316358 DOI: 10.1186/s12967-024-05552-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Although immune checkpoint inhibitors (anti-PD-1 antibody, anti-PD-L1 antibody, and anti-CTLA-4 antibody) have displayed considerable success in the treatment of malignant tumors, the therapeutic effect is still unsatisfactory for a portion of patients. Therefore, it is imperative to develop strategies to enhance the effect of these ICIs. Increasing evidence strongly suggests that the key to this issue is to transform the tumor immune microenvironment from a state of no or low immune infiltration to a state of high immune infiltration and enhance the tumor cell-killing effect of T cells. Therefore, some combination strategies have been proposed and this review appraise a summary of 39 strategies aiming at enhancing the effectiveness of ICIs, which comprise combining 10 clinical approaches and 29 foundational research strategies. Moreover, this review improves the comprehensive understanding of combination therapy with ICIs and inspires novel ideas for tumor immunotherapy.
Collapse
Affiliation(s)
- Xin Su
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Jian Li
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Xiao Xu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Youbao Ye
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Cailiu Wang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Guanglong Pang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Wenxiu Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Ang Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Changchun Zhao
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Xiangyong Hao
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China.
| |
Collapse
|
35
|
Xu H, Russell SN, Steiner K, O'Neill E, Jones KI. Targeting PI3K-gamma in myeloid driven tumour immune suppression: a systematic review and meta-analysis of the preclinical literature. Cancer Immunol Immunother 2024; 73:204. [PMID: 39105848 PMCID: PMC11303654 DOI: 10.1007/s00262-024-03779-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 07/11/2024] [Indexed: 08/07/2024]
Abstract
The intricate interplay between immune and stromal cells within the tumour microenvironment (TME) significantly influences tumour progression. Myeloid cells, including tumour-associated macrophages (TAMs), neutrophils (TANs), and myeloid-derived suppressor cells (MDSCs), contribute to immune suppression in the TME (Nakamura and Smyth in Cell Mol Immunol 17(1):1-12 (2020). https://doi.org/10.1038/s41423-019-0306-1 ; DeNardo and Ruffell in Nat Rev Immunol 19(6):369-382 (2019). https://doi.org/10.1038/s41577-019-0127-6 ). This poses a significant challenge for novel immunotherapeutics that rely on host immunity to exert their effect. This systematic review explores the preclinical evidence surrounding the inhibition of phosphoinositide 3-kinase gamma (PI3Kγ) as a strategy to reverse myeloid-driven immune suppression in solid tumours. EMBASE, MEDLINE, and PubMed databases were searched on 6 October 2022 using keyword and subject heading terms to capture relevant studies. The studies, focusing on PI3Kγ inhibition in animal models, were subjected to predefined inclusion and exclusion criteria. Extracted data included tumour growth kinetics, survival endpoints, and immunological responses which were meta-analysed. PRISMA and MOOSE guidelines were followed. A total of 36 studies covering 73 animal models were included in the review and meta-analysis. Tumour models covered breast, colorectal, lung, skin, pancreas, brain, liver, prostate, head and neck, soft tissue, gastric, and oral cancer. The predominant PI3Kγ inhibitors were IPI-549 and TG100-115, demonstrating favourable specificity for the gamma isoform. Combination therapies, often involving chemotherapy, radiotherapy, immune checkpoint inhibitors, biological agents, or vaccines, were explored in 81% of studies. Analysis of tumour growth kinetics revealed a statistically significant though heterogeneous response to PI3Kγ monotherapy, whereas the tumour growth in combination treated groups were more consistently reduced. Survival analysis showed a pronounced increase in median overall survival with combination therapy. This systematic review provides a comprehensive analysis of preclinical studies investigating PI3Kγ inhibition in myeloid-driven tumour immune suppression. The identified studies underscore the potential of PI3Kγ inhibition in reshaping the TME by modulating myeloid cell functions. The combination of PI3Kγ inhibition with other therapeutic modalities demonstrated enhanced antitumour effects, suggesting a synergistic approach to overcome immune suppression. These findings support the potential of PI3Kγ-targeted therapies, particularly in combination regimens, as a promising avenue for future clinical exploration in diverse solid tumour types.
Collapse
Affiliation(s)
- Haonan Xu
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | - Eric O'Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | - Keaton Ian Jones
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
36
|
Liu J, Wu J, Chen T, Yang B, Liu X, Xi J, Zhang Z, Gao Y, Li Z. Enhancing X-Ray Sensitization with Multifunctional Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400954. [PMID: 38676336 DOI: 10.1002/smll.202400954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/30/2024] [Indexed: 04/28/2024]
Abstract
In the progression of X-ray-based radiotherapy for the treatment of cancer, the incorporation of nanoparticles (NPs) has a transformative impact. This study investigates the potential of NPs, particularly those comprised of high atomic number elements, as radiosensitizers. This aims to optimize localized radiation doses within tumors, thereby maximizing therapeutic efficacy while preserving surrounding tissues. The multifaceted applications of NPs in radiotherapy encompass collaborative interactions with chemotherapeutic, immunotherapeutic, and targeted pharmaceuticals, along with contributions to photodynamic/photothermal therapy, imaging enhancement, and the integration of artificial intelligence technology. Despite promising preclinical outcomes, the paper acknowledges challenges in the clinical translation of these findings. The conclusion maintains an optimistic stance, emphasizing ongoing trials and technological advancements that bolster personalized treatment approaches. The paper advocates for continuous research and clinical validation, envisioning the integration of NPs as a revolutionary paradigm in cancer therapy, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, 410011, China
| | - JunYong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, 410011, China
| | - Taili Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Bin Yang
- Department of Orthopedics, Shaodong People's Hospital, Shaoyang, Hunan Province, 422800, China
| | - XiangPing Liu
- Department of Neurology, Shaodong People's Hospital, Shaoyang, Hunan Province, 422800, China
| | - Jing Xi
- Department of Nephrology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changde, Hunan Province, 415000, China
| | - Ziyang Zhang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 119276, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
| | - Yawen Gao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, 410011, China
| | - ZhiHong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, 410011, China
| |
Collapse
|
37
|
Chen G, Lin L, Mai Z, Tang Y, Zhang Q, Chen G, Li Z, Zhang J, Wang Y, Yang Y, Yu Z. Carrier-Free Photodynamic Bioregulators Inhibiting Lactic Acid Efflux Combined with Immune Checkpoint Blockade for Triple-Negative Breast Cancer Immunotherapy. ACS NANO 2024. [PMID: 39034461 DOI: 10.1021/acsnano.4c07213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Abnormal tumor metabolism creates a complex tumor immune microenvironment that plays a dominant role in the metastasis of triple-negative breast cancer (TNBC). TNBC is insensitive to immune checkpoint blockade (ICB) therapy because of insufficient cytotoxic T lymphocyte (CTL) infiltration and a hyper-lactic acid-suppressive immune microenvironment caused by abnormal glycolysis. Herein, we propose an amplified strategy based on lactic acid regulation to reprogram the immunosuppressive tumor microenvironment (ITM) and combine it with ICB therapy to achieve enhanced antitumor immunotherapy effects. Specifically, we constructed CASN, a carrier-free photodynamic bioregulator, through the self-assembly of the photosensitizer Chlorin e6 and monocarboxylate transporter 1 (MCT1) inhibitor AZD3965. CASN exhibited a uniform structure, good stability, and drug accumulation at the tumor site. CASN-mediated photodynamic therapy following laser irradiation inhibited primary tumor growth and induced immunogenic cell death. Furthermore, CASN reduced lactic acid-mediated regulatory T cell generation and M2 tumor-associated macrophage polarization by blocking MCT1-mediated lactic acid efflux to attenuate immune suppression, inducing the recruitment and activation of CTLs. Ultimately, CASN-mediated immunopotentiation combined with ICB therapy considerably strengthened tumor immunotherapy and effectively inhibited tumor growth and metastasis of TNBC. This synergistic amplification strategy overcomes the limitations of an acidic ITM and presents a potential clinical treatment option for metastatic tumors.
Collapse
Affiliation(s)
- Guimei Chen
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ling Lin
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ziyi Mai
- Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Yan Tang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qiaoling Zhang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Gui Chen
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zibo Li
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiasi Zhang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
| | - Yongxia Wang
- Department of Galactophore, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523058, China
| | - Yuanyuan Yang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
| | - Zhiqiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
| |
Collapse
|
38
|
Varpe P, Joga R, Aglave G, Vasu P, Yerram S, Bellapu KK, Srivastava S, Kumar S. Esterase responsive release of anti-cancer agents from conjugated lipid nanocarrier and the regulatory considerations. Pharm Pat Anal 2024; 13:31-43. [PMID: 39324857 PMCID: PMC11449025 DOI: 10.1080/20468954.2024.2347796] [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: 10/19/2023] [Accepted: 02/20/2024] [Indexed: 09/27/2024]
Abstract
The release of active agents in tumors rather than normal tissues, limits systemic exposure and toxicities. Targeting over-expressed esterase enzyme in the tumor microenvironment can selectively release immune-active agents like Programmed Death-1 (PD-1) and PD-1 ligand inhibitors from ester-sensitive lipid nanocarriers, offering a novel approach compared with conventional therapies. PD-1 and PD-L1 association cause T-cell inactivation, whereas blocking their association improves their cytotoxic mechanism. The patent application US2022/0080051-A1 discloses a novel immune-active agent conjugated with lipid to form a nanocarrier for esterase-sensitive release. These nanocarriers selectively enter leaky vasculature of tumors through enhanced permeability and retention effect, undergo ester cleavage to release agents, and are reported to increase bioavailability by 24 times. Further, with other agents or alone it achieves targeted synergistic cancer therapy. Also, the current patent spotlight delves into the crucial formulation considerations necessary for obtaining successful approval of lipidic nano products from relevant regulatory authorities.
Collapse
Affiliation(s)
- Priya Varpe
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Ramesh Joga
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Gayatri Aglave
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Pavan Vasu
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Sravani Yerram
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Kiran Kumar Bellapu
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana , 500037, India
| | - Sandeep Kumar
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
- Department of Pharmaceutics, Nims Institute of Pharmacy, Nims University Rajasthan, Jaipur, Rajasthan 303121, India
| |
Collapse
|
39
|
Yang C, Jiang Y, Zhang K, Zhu X, Li J, Yu H, Chen J, Gu X, Gan Z, Yu Q. Photodynamic Therapy Derived Personalized Whole Cell Tumor Vaccine Prevents Postsurgery Tumor Recurrence and Metastasis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308456. [PMID: 38342675 DOI: 10.1002/smll.202308456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/03/2024] [Indexed: 02/13/2024]
Abstract
In order to avoid the time-consuming and laborious identification of tumor-specific antigens (TSAs) during the traditional vaccine fabrication process, a versatile photodynamic therapy (PDT)-based method is developed to construct a whole-tumor antigen tumor vaccine (TV) from surgically resected tumor tissues for personalized immunotherapy. Mucoadhesive nanoparticles containing small-molecular photosensitizer are fabricated and directly co-incubated with suspended tumor cells obtained after cytoreduction surgery. After irradiation with a 405 nm laser, potent immunogenic cell death of cancer cells could be induced. Along with the release of TSAs, the as-prepared TV could activate safe and robust tumor-specific immune responses, leading to efficient suppression of postsurgery tumor recurrence and metastasis. The as-prepared TV cannot only be applied alone through various administration routes but also synergize with immunoadjuvant, chemotherapeutics, and immune checkpoint blockers to exert more potent immune responses. This work provides an alternative way to promote the clinical translation of PDT, which is generally restricted by the limited penetration of light. Moreover, the versatile strategy of vaccine fabrication also facilitates the clinical application of personalized whole-cell tumor vaccines.
Collapse
Affiliation(s)
- Chunyu Yang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yitong Jiang
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Kaixin Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xianqi Zhu
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jianlin Li
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Haiwang Yu
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jiawei Chen
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xinggui Gu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- Beijing National Laboratory for Molecular Sciences, Beijing, 100190, China
| | - Zhihua Gan
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qingsong Yu
- The State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
40
|
Gu J, Cheng D, Li H, Yu T, Zhang Z, Liu Y, Wang X, Lu X, Li J. Radioactive hybrid semiconducting polymer nanoparticles for imaging-guided tri-modal therapy of breast cancer. J Mater Chem B 2024; 12:6091-6101. [PMID: 38828732 DOI: 10.1039/d4tb00834k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Due to the rapid progression and aggressive metastasis of breast cancer, its diagnosis and treatment remain a great challenge. The simultaneous inhibition of tumor growth and metastasis is necessary for breast cancer to obtain ideal therapeutic outcomes. We herein report the development of radioactive hybrid semiconducting polymer nanoparticles (SPNH) for imaging-guided tri-modal therapy of breast cancer. Two semiconducting polymers are used to form SPNH with a diameter of around 60 nm via nano-coprecipitation and they are also labeled with iodine-131 (131I) to enhance the imaging functions. The formed SPNH show good radiolabeling stability and excellent photodynamic and photothermal effects under 808 nm laser irradiation to produce singlet oxygen (1O2) and heat. Moreover, SPNH can generate 1O2 with ultrasound irradiation via their sonodynamic properties. After intravenous tail vein injection, SPNH can effectively accumulate in the subcutaneous 4T1 tumors of living mice as verified via fluorescence and single photon emission computed tomography (SPECT) imaging. With the irradiation of tumors using an 808 nm laser and US, SPNH mediate photodynamic therapy (PDT), photothermal therapy (PTT) and sonodynamic therapy (SDT) to kill tumor cells. Such a tri-modal therapy leads to an improved efficacy in inhibiting tumor growth and suppressing tumor metastasis compared to the sole SDT and combinational PDT-PTT. This study thus demonstrates the applications of SPNH to diagnose tumors and combine different therapies for effective breast cancer treatment.
Collapse
Affiliation(s)
- Junhao Gu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Danling Cheng
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Haiyan Li
- Department of Nuclear Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou 225001, P. R. China.
| | - Tao Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Zhenghe Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Yue Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Xiaoying Wang
- Office of Hospital Infection and Disease Control and Prevention, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China.
| | - Xia Lu
- Department of Nuclear Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou 225001, P. R. China.
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China.
| |
Collapse
|
41
|
Guo Z, Ye J, Cheng X, Wang T, Zhang Y, Yang K, Du S, Li P. Nanodrug Delivery Systems in Antitumor Immunotherapy. Biomater Res 2024; 28:0015. [PMID: 38840653 PMCID: PMC11045275 DOI: 10.34133/bmr.0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/26/2024] [Indexed: 06/07/2024] Open
Abstract
Cancer has become one of the most important factors threatening human health, and the global cancer burden has been increasing rapidly. Immunotherapy has become another clinical research hotspot after surgery, chemotherapy, and radiotherapy because of its high efficiency and tumor metastasis prevention. However, problems such as lower immune response rate and immune-related adverse reaction in the clinical application of immunotherapy need to be urgently solved. With the development of nanodrug delivery systems, various nanocarrier materials have been used in the research of antitumor immunotherapy with encouraging therapeutic results. In this review, we mainly summarized the combination of nanodrug delivery systems and immunotherapy from the following 4 aspects: (a) nanodrug delivery systems combined with cytokine therapy to improve cytokines delivery in vivo; (b) nanodrug delivery systems provided a suitable platform for the combination of immune checkpoint blockade therapy with other tumor treatments; (c) nanodrug delivery systems helped deliver antigens and adjuvants for tumor vaccines to enhance immune effects; and (d) nanodrug delivery systems improved tumor treatment efficiency and reduced toxicity for adoptive cell therapy. Nanomaterials chosen by researchers to construct nanodrug delivery systems and their function were also introduced in detail. Finally, we discussed the current challenges and future prospects in combining nanodrug delivery systems with immunotherapy.
Collapse
Affiliation(s)
- Zishuo Guo
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jinhong Ye
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xuehao Cheng
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Tieshan Wang
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yi Zhang
- YiDu Central Hospital of Weifang, Weifang, Shandong 262500, China
| | - Kaili Yang
- Beijing University of Chinese Medicine, Beijing 102488, China
| | | | - Pengyue Li
- Address correspondence to: (P.L.); (S.D.)
| |
Collapse
|
42
|
Geng S, Guo P, Wang J, Zhang Y, Shi Y, Li X, Cao M, Song Y, Zhang H, Zhang Z, Zhang K, Song H, Shi J, Liu J. Ultrasensitive Optical Detection and Elimination of Residual Microtumors with a Postoperative Implantable Hydrogel Sensor for Preventing Cancer Recurrence. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307923. [PMID: 38174840 DOI: 10.1002/adma.202307923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/16/2023] [Indexed: 01/05/2024]
Abstract
In vivo optical imaging of trace biomarkers in residual microtumors holds significant promise for cancer prognosis but poses a formidable challenge. Here, a novel hydrogel sensor is designed for ultrasensitive and specific imaging of the elusive biomarker. This hydrogel sensor seamlessly integrates a molecular beacon nanoprobe with fibroblasts, offering both high tissue retention capability and an impressive signal-to-noise ratio for imaging. Signal amplification is accomplished through exonuclease I-mediated biomarker recycling. The resulting hydrogel sensor sensitively detects the biomarker carcinoembryonic antigen with a detection limit of 1.8 pg mL-1 in test tubes. Moreover, it successfully identifies residual cancer nodules with a median diameter of less than 2 mm in mice bearing partially removed primary triple-negative breast carcinomas (4T1). Notably, this hydrogel sensor is proven effective for the sensitive diagnosis of invasive tumors in post-surgical mice with infiltrating 4T1 cells, leveraging the role of fibroblasts in locally enriching tumor cells. Furthermore, the residual microtumor is rapidly photothermal ablation by polydopamine-based nanoprobe under the guidance of visualization, achieving ≈100% suppression of tumor recurrence and lung metastasis. This work offers a promising alternative strategy for visually detecting residual microtumors, potentially enhancing the prognosis of cancer patients following surgical interventions.
Collapse
Affiliation(s)
- Shizhen Geng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Pengke Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jing Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yunya Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yaru Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xinling Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Mengnian Cao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yutong Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hongling Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Haiwei Song
- Department of Biochemistry, National University of Singapore, SingaporeCity, 138673, Singapore
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| |
Collapse
|
43
|
Wang M, Xue W, Yuan H, Wang Z, Yu L. Nano-Drug Delivery Systems Targeting CAFs: A Promising Treatment for Pancreatic Cancer. Int J Nanomedicine 2024; 19:2823-2849. [PMID: 38525013 PMCID: PMC10959015 DOI: 10.2147/ijn.s451151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/06/2024] [Indexed: 03/26/2024] Open
Abstract
Currently, pancreatic cancer (PC) is one of the most lethal malignant tumors. PC is typically diagnosed at a late stage, exhibits a poor response to conventional treatment, and has a bleak prognosis. Unfortunately, PC's survival rate has not significantly improved since the 1960s. Cancer-associated fibroblasts (CAFs) are a key component of the pancreatic tumor microenvironment (TME). They play a vital role in maintaining the extracellular matrix and facilitating the intricate communication between cancer cells and infiltrated immune cells. Exploring therapeutic approaches targeting CAFs may reverse the current landscape of PC therapy. In recent years, nano-drug delivery systems have evolved rapidly and have been able to accurately target and precisely release drugs with little or no toxicity to the whole body. In this review, we will comprehensively discuss the origin, heterogeneity, potential targets, and recent advances in the nano-drug delivery system of CAFs in PC. We will also propose a novel integrated treatment regimen that utilizes a nano-drug delivery system to target CAFs in PC, combined with radiotherapy and immunotherapy. Additionally, we will address the challenges that this regimen currently faces.
Collapse
Affiliation(s)
- Mingjie Wang
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Wenxiang Xue
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Hanghang Yuan
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Lei Yu
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
44
|
Lin J, Ma Z, Zuo W, Zhu M. Enhancing Targeted Photodynamic Therapy: Star-Shaped Glycopolymeric Photosensitizers for Improved Selectivity and Efficacy. Biomacromolecules 2024; 25:1950-1958. [PMID: 38334281 DOI: 10.1021/acs.biomac.3c01378] [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: 02/10/2024]
Abstract
Targeted photodynamic therapy (PDT) offers advantages over nontargeted approaches, including improved selectivity, efficacy, and reduced side effects. This study developed star-shaped glycopolymeric photosensitizers using porphyrin-based initiators via ATRP. Incorporating a porphyrin core gave the polymers fluorescence and ROS generation, while adding fructose improved solubility and targeting capabilities. The photosensitizers had high light absorption, singlet oxygen production, specificity, low dark toxicity, and biocompatibility. The glycopolymers with longer sugar arms and higher density showed better uptake on MCF-7 and MDA-MB-468 cells compared to HeLa cells, indicating enhanced targeting capabilities. Inhibition of endocytosis confirmed the importance of the GLUT5 receptor. The resulting polymers exhibited good cytocompatibility under dark conditions and satisfactory PDT under light irradiation. Interestingly, the polymers containing fructose have a GLUT5-dependent elimination effect on the MCF-7 and MDA-MB-468 cells. The intracellular ROS production followed a similar pattern, indicating that the fructose polymer exhibits specific targeting toward cells with GLUT5 receptors.
Collapse
Affiliation(s)
- Jiahui Lin
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China
| | - Zhiyuan Ma
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China
| | - Weiwei Zuo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China
| |
Collapse
|
45
|
Zhu X, Zheng W, Wang X, Li Z, Shen X, Chen Q, Lu Y, Chen K, Ai S, Zhu Y, Guan W, Yao S, Liu S. Enhanced Photodynamic Therapy Synergizing with Inhibition of Tumor Neutrophil Ferroptosis Boosts Anti-PD-1 Therapy of Gastric Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307870. [PMID: 38233204 DOI: 10.1002/advs.202307870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/19/2023] [Indexed: 01/19/2024]
Abstract
For tumor treatment, the ultimate goal in tumor therapy is to eliminate the primary tumor, manage potential metastases, and trigger an antitumor immune response, resulting in the complete clearance of all malignant cells. Tumor microenvironment (TME) refers to the local biological environment of solid tumors and has increasingly become an attractive target for cancer therapy. Neutrophils within TME of gastric cancer (GC) spontaneously undergo ferroptosis, and this process releases oxidized lipids that limit T cell activity. Enhanced photodynamic therapy (PDT) mediated by di-iodinated IR780 (Icy7) significantly increases the production of reactive oxygen species (ROS). Meanwhile, neutrophil ferroptosis can be triggered by increased ROS generation in the TME. In this study, a liposome encapsulating both ferroptosis inhibitor Liproxstatin-1 and modified photosensitizer Icy7, denoted LLI, significantly inhibits tumor growth of GC. LLI internalizes into MFC cells to generate ROS causing immunogenic cell death (ICD). Simultaneously, liposome-deliver Liproxstatin-1 effectively inhibits the ferroptosis of tumor neutrophils. LLI-based immunogenic PDT and neutrophil-targeting immunotherapy synergistically boost the anti-PD-1 treatment to elicit potent TME and systemic antitumor immune response with abscopal effects. In conclusion, LLI holds great potential for GC immunotherapy.
Collapse
Affiliation(s)
- Xudong Zhu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Wenxuan Zheng
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xingzhou Wang
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Zhiyan Li
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xiaofei Shen
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Qi Chen
- China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210008, China
| | - Yanjun Lu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Kai Chen
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Shichao Ai
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Wenxian Guan
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Shankun Yao
- State Key Laboratory of Coordination Chemistry, Coordination Chemistry Institute, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Song Liu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| |
Collapse
|
46
|
Zhang Z, Yu D, Sui D, Shi M, Wang K, Zhang Y, Ji Y. Manganese Dioxide Nanoplatform with a Hollow Rhombic Dodecahedron Morphology for Drug Delivery. ACS APPLIED BIO MATERIALS 2024; 7:1169-1178. [PMID: 38253011 DOI: 10.1021/acsabm.3c01068] [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: 01/24/2024]
Abstract
Manganese dioxide (MnO2) is considered as a promising drug carrier material suitable for the tumor microenvironment while lacking conducive structures for drug loading. Herein, we construct a MnO2 nanoplatform with a hollow rhombic dodecahedral morphology for drug delivery. In this work, we obtained zeolitic imidazolate framework nanoparticles (ZIF-90 NPs) via a coordination reaction. Furthermore, the drug-loading nanoparticles (ZIF-90/DOX NPs) were obtained by Schiff's base reaction and then selected as a sacrificial template to obtain the hollow nanoplatforms (ZIF-90@MnO2 NPs). Moreover, the photothermal effect and multiresponsive drug release behaviors were revealed by loading the photothermal agent IR-820 and the anticancer drug doxorubicin hydrochloride (DOX). Our study demonstrates that the ZIF-90@MnO2 NPs loaded with photosensitizers exhibited excellent photothermal conversion performance. Benefiting from the hollow structure and redox activity, remarkable drug loading and release performances of ZIF-90@MnO2 NPs were achieved. It is shown that ZIF-90@MnO2 NPs achieved a satisfactory drug-loading efficiency (up to ca. 69.7%) for DOX. More promisingly, the ZIF-90@MnO2 NPs exhibited significant glutathione (GSH)/pH-responsive drug release and degradation performances. Overall, this work highlights the potential of controlled drug release of nanocarriers and offers unique insights into the design of nanocarriers with hollow structures.
Collapse
Affiliation(s)
- Zheng Zhang
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Danlu Yu
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Dan Sui
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Miaomiao Shi
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Kangjun Wang
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Yajing Zhang
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Yuanhui Ji
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
47
|
Zhang Z, Liang X, Yang X, Liu Y, Zhou X, Li C. Advances in Nanodelivery Systems Based on Metabolism Reprogramming Strategies for Enhanced Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6689-6708. [PMID: 38302434 DOI: 10.1021/acsami.3c15686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Tumor development and metastasis are closely related to the complexity of the metabolism network. Recently, metabolism reprogramming strategies have attracted much attention in tumor metabolism therapy. Although there is preliminary success of metabolism therapy agents, their therapeutic effects have been restricted by the effective reaching of the tumor sites of drugs. Nanodelivery systems with unique physical properties and elaborate designs can specifically deliver to the tumors. In this review, we first summarize the research progress of nanodelivery systems based on tumor metabolism reprogramming strategies to enhance therapies by depleting glucose, inhibiting glycolysis, depleting lactic acid, inhibiting lipid metabolism, depleting glutamine and glutathione, and disrupting metal metabolisms combined with other therapies, including chemotherapy, radiotherapy, photodynamic therapy, etc. We further discuss in detail the advantages of nanodelivery systems based on tumor metabolism reprogramming strategies for tumor therapy. As well as the opportunities and challenges for integrating nanodelivery systems into tumor metabolism therapy, we analyze the outlook for these emerging areas. This review is expected to improve our understanding of modulating tumor metabolisms for enhanced therapy.
Collapse
Affiliation(s)
- Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiangyu Zhou
- Department of Thyroid and Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
48
|
Palvai S, Kpeglo D, Newham G, Peyman SA, Evans SD, Ong ZY. Free-Standing Hierarchically Porous Silica Nanoparticle Superstructures: Bridging the Nano- to Microscale for Tailorable Delivery of Small and Large Therapeutics. ACS APPLIED MATERIALS & INTERFACES 2024; 16:5568-5581. [PMID: 38270578 PMCID: PMC10859928 DOI: 10.1021/acsami.3c16463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/13/2023] [Accepted: 01/08/2024] [Indexed: 01/26/2024]
Abstract
Nanoscale colloidal self-assembly is an exciting approach to yield superstructures with properties distinct from those of individual nanoparticles. However, the bottom-up self-assembly of 3D nanoparticle superstructures typically requires extensive chemical functionalization, harsh conditions, and a long preparation time, which are undesirable for biomedical applications. Here, we report the directional freezing of porous silica nanoparticles (PSiNPs) as a simple and versatile technique to create anisotropic 3D superstructures with hierarchical porosity afforded by microporous PSiNPs and newly generated meso- and macropores between the PSiNPs. By varying the PSiNP building block size, the interparticle pore sizes can be readily tuned. The newly created hierarchical pores greatly augment the loading of a small molecule-anticancer drug, doxorubicin (Dox), and a large macromolecule, lysozyme (Lyz). Importantly, Dox loading into both the micro- and meso/macropores of the nanoparticle assemblies not only gave a pore size-dependent drug release but also significantly extended the drug release to 25 days compared to a much shorter 7 or 11 day drug release from Dox loaded into either the micro- or meso/macropores only. Moreover, a unique temporal drug release profile, with a higher and faster release of Lyz from the larger interparticle macropores than Dox from the smaller PSiNP micropores, was observed. Finally, the formulation of the Dox-loaded superstructures within a composite hydrogel induces prolonged growth inhibition in a 3D spheroid model of pancreatic ductal adenocarcinoma. This study presents a facile modular approach for the rapid assembly of drug-loaded superstructures in fully aqueous environments and demonstrates their potential as highly tailorable and sustained delivery systems for diverse therapeutics.
Collapse
Affiliation(s)
- Sandeep Palvai
- School
of Physics and Astronomy, University of
Leeds, Leeds LS2 9JT, U.K.
| | - Delanyo Kpeglo
- School
of Physics and Astronomy, University of
Leeds, Leeds LS2 9JT, U.K.
| | - George Newham
- School
of Physics and Astronomy, University of
Leeds, Leeds LS2 9JT, U.K.
| | - Sally A. Peyman
- School
of Physics and Astronomy, University of
Leeds, Leeds LS2 9JT, U.K.
- Leeds
Institute of Medical Research at St James, School of Medicine, University of Leeds, Leeds LS2 9JT, U.K.
| | - Stephen D. Evans
- School
of Physics and Astronomy, University of
Leeds, Leeds LS2 9JT, U.K.
| | - Zhan Yuin Ong
- School
of Physics and Astronomy, University of
Leeds, Leeds LS2 9JT, U.K.
- Leeds
Institute of Medical Research at St James, School of Medicine, University of Leeds, Leeds LS2 9JT, U.K.
| |
Collapse
|
49
|
Wang F, Dong G, Ding M, Yu N, Sheng C, Li J. Dual-Programmable Semiconducting Polymer NanoPROTACs for Deep-Tissue Sonodynamic-Ferroptosis Activatable Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306378. [PMID: 37817359 DOI: 10.1002/smll.202306378] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/14/2023] [Indexed: 10/12/2023]
Abstract
Proteolysis-targeting chimeras (PROTACs) can provide promising opportunities for cancer treatment, while precise regulation of their activities remains challenging to achieve effective and safe therapeutic outcomes. A semiconducting polymer nanoPROTAC (SPNFeP ) is reported that can achieve ultrasound (US) and tumor microenvironment dual-programmable PROTAC activity for deep-tissue sonodynamic-ferroptosis activatable immunotherapy. SPNFeP is formed through a nano-precipitation of a sonodynamic semiconducting polymer, a ferroptosis inducer, and a newly synthesized PROTAC molecule. The semiconducting polymers work as sonosensitizers to produce singlet oxygen (1 O2 ) via sonodynamic effect under US irradiation, and ferroptosis inducers react with intratumoral hydrogen peroxide (H2 O2 ) to generate hydroxyl radical (·OH). Such a dual-programmable reactive oxygen species (ROS) generation not only triggers ferroptosis and immunogenic cell death (ICD), but also induces on-demand activatable delivery of PROTAC molecules into tumor sites. The effectively activated nanoPROTACs degrade nicotinamide phosphoribosyl transferase (NAMPT) to suppress tumor infiltration of myeloid-derived suppressive cells (MDSCs), thus promoting antitumor immunity. In such a way, SPNFeP mediates sonodynamic-ferroptosis activatable immunotherapy for entirely inhibiting tumor growths in both subcutaneous and 2-cm tissue-covered deep tumor mouse models. This study presents a dual-programmable activatable strategy based on PROTACs for effective and precise cancer combinational therapy.
Collapse
Affiliation(s)
- Fengshuo Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Guoqiang Dong
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Mengbin Ding
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Ningyue Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Chunquan Sheng
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
50
|
Chen S, Luo Y, He Y, Li M, Liu Y, Zhou X, Hou J, Zhou S. In-situ-sprayed therapeutic hydrogel for oxygen-actuated Janus regulation of postsurgical tumor recurrence/metastasis and wound healing. Nat Commun 2024; 15:814. [PMID: 38280861 PMCID: PMC10821930 DOI: 10.1038/s41467-024-45072-x] [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: 06/30/2023] [Accepted: 01/12/2024] [Indexed: 01/29/2024] Open
Abstract
Surgery is the mainstay of treatment modality for malignant melanoma. However, the deteriorative hypoxic microenvironment after surgery is recognized as a stemming cause for tumor recurrence/metastasis and delayed wound healing. Here we design and construct a sprayable therapeutic hydrogel (HIL@Z/P/H) encapsulating tumor-targeted nanodrug and photosynthetic cyanobacteria (PCC 7942) to prevent tumor recurrence/metastasis while promote wound healing. In a postsurgical B16F10 melanoma model in female mice, the nanodrug can disrupt cellular redox homeostasis via the photodynamic therapy-induced cascade reactions within tumor cells. Besides, the photosynthetically generated O2 by PCC 7942 can not only potentiate the oxidative stress-triggered cell death to prevent local recurrence of residual tumor cells, but also block the signaling pathway of hypoxia-inducible factor 1α to inhibit their distant metastasis. Furthermore, the long-lasting O2 supply and PCC 7942-secreted extracellular vesicles can jointly promote angiogenesis and accelerate the wound healing process. Taken together, the developed HIL@Z/P/H capable of preventing tumor recurrence/metastasis while promoting wound healing shows great application potential for postsurgical cancer therapy.
Collapse
Affiliation(s)
- Shuiling Chen
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yang Luo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yang He
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Ming Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yongjian Liu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xishen Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jianwen Hou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|