1
|
Hu Z, Zhou X, Zhang W, Zhang L, Li L, Gao Y, Wang C. Photothermal amplified multizyme activity for synergistic photothermal-catalytic tumor therapy. J Colloid Interface Sci 2025; 679:375-383. [PMID: 39366266 DOI: 10.1016/j.jcis.2024.09.223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 10/06/2024]
Abstract
Nano-enzymatic catalytic therapy has been widely explored as a promising tumor therapeutic method with specific responsiveness to the tumor microenvironment (TME). However, the inherent lower and simplex catalytic efficiency impairs their anti-tumor efficacy. Therefore, developing novel nanozymes with relatively high and multiple catalytic characteristics, simultaneously enhancing the enzyme-like activity of nanozymes using the proper method, photothermal promoted catalytic property, is a reliable way. In this paper, we report a manganese oxide/nitrogen-doped carbon composite nanoparticles (MnO-N/C NPs) with multi-enzyme mimetic activity and photothermal conversional effect. The peroxidase (POD)-like/oxidase (OXD)-like/catalase (CAT)-like activity of MnO-N/C nanozymes was accelerated upon exposure to an 808 nm NIR laser. In vitro and in vivo results proved that the MnO-N/C NPs shown excellent magnetic resonance imaging (MRI) guided synergistic photothermal-enhanced catalytic treatment and photothermal therapy of liver cancer. The photothermal enhanced multi-enzyme activity maximizes the efficacy of catalytic and photothermal therapy while reducing harm to healthy cells, thereby offering valuable insights for the development of next-generation photothermal nanozymes to enhance tumor therapy.
Collapse
Affiliation(s)
- Zhichao Hu
- Faculty of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin 130024, PR China
| | - Xue Zhou
- Faculty of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin 130024, PR China
| | - Wei Zhang
- Faculty of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin 130024, PR China
| | - Lingyu Zhang
- Faculty of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin 130024, PR China
| | - Lu Li
- Faculty of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin 130024, PR China
| | - Ying Gao
- Department of Stomatology, No. 964 Hospital, Changchun, Jilin 130021, PR China.
| | - Chungang Wang
- Faculty of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin 130024, PR China.
| |
Collapse
|
2
|
Cun JE, He Z, Fan X, Pan Q, Luo K, He B, Pu Y. Copper-Based Bio-Coordination Nanoparticle for Enhanced Pyroptosis-Cuproptosis Cancer Immunotherapy through Redox Modulation and Glycolysis Inhibition. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2409875. [PMID: 39757406 DOI: 10.1002/smll.202409875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/19/2024] [Indexed: 01/07/2025]
Abstract
Copper-based nanoparticles have garnered significant interest in cancer therapy due to their ability to induce oxidative stress and cuproptosis in cancer cells. However, their antitumor effectiveness is constrained by the dynamic redox balance and the metabolic shift between oxidative phosphorylation and glycolysis. Here, a polydopamine-coated copper-α-ketoglutaric acid (α-KG) coordination polymer nanoparticle (CKPP) is designed for combined pyroptosis-cuproptosis cancer immunotherapy by amplifying reactive oxygen species (ROS) production and regulating cellular metabolism. The intracellular redox imbalance is achieved through the synergistic effects of α-KG-induced mitochondrial metabolic reprogramming, photothermally enhanced superoxide dismutase-like activity of polydopamine, and glutathione depletion by copper ions. The multifaceted redox modulation results in a substantial increase in intracellular ROS levels, triggering oxidative stress and subsequent pyroptosis in cancer cells. Furthermore, α-KG shifts cellular metabolism from glycolysis to oxidative phosphorylation, thereby enhancing cuproptosis induced by copper ions. The combination of ROS dyshomeostasis and glycolysis inhibition results in a potent enhancement of pyroptosis-cuproptosis-mediated cancer therapy. In a murine model of colorectal cancer, CKPP exhibited a remarkable anticancer effect, achieving a tumor inhibition rate of 96.3% and complete tumor eradication in two out of five cases. Overall, this bio-engineered metal-organic nanocomposite demonstrates significant potential for treating cancer through combined pyroptosis-cuproptosis cancer immunotherapy.
Collapse
Affiliation(s)
- Ju-E Cun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Med-X Center for Materials, Sichuan University, Chengdu, 610064, China
| | - Ziyun He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Med-X Center for Materials, Sichuan University, Chengdu, 610064, China
| | - Xi Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Med-X Center for Materials, Sichuan University, Chengdu, 610064, China
| | - Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu, 610106, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu, 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Med-X Center for Materials, Sichuan University, Chengdu, 610064, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Med-X Center for Materials, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
3
|
Yang Y, Xu Q, Gu W, Nan K, Chen S, Wang S, Zhang J, Zhao Q. Oxidative stress-augmented Cu-doped hollow mesoporous carbon nanozyme for photothermal/photodynamic synergistic therapy. J Colloid Interface Sci 2024; 683:910-925. [PMID: 39709766 DOI: 10.1016/j.jcis.2024.12.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Photodynamic therapy (PDT) has witnessed remarkable progress in recent years owing to its specific properties. Given that the antioxidation system of tumor microenvironment (TME) adversely affects treatment outcomes, powerful TME modulators can significantly resolve the limitation of PDT. Herein, we developed a PEG-modified Cu2+-doped hollow mesoporous carbon nanozyme (CHC-PEG) and loaded insoluble photosensitizer IR780 into its pores and cavities to construct the multifunctional nano-system IR780/CHCP. CHC-PEG nanozyme could perform photothermal therapy (PTT) effect and protect IR780 from aggregation-caused quenching (ACQ) effect, while exerting peroxidase (POD)-mimetic activity and the ability of consuming glutathione (GSH) to achieve oxidative stress-augmented PDT effect. When exposed to near-infrared (NIR) light, IR780 was stimulated to produce singlet oxygen (1O2) and CHC-PEG could increase the temperature of TME to exert stronger POD-mimetic activity for producing more hydroxyl radicals (OH), therefore the IR780/CHCP nano-system exhibited remarkable tumor growth inhibition. Benefited by the enhanced synergistic effect, IR780/CHCP exhibited remarkable in vivo tumor growth inhibition, with the tumor inhibition rate of 93 %, and had no significant effect on major organs. Above all, IR780/CHCP could resist the antioxidant system in TME to enhance the level of oxidative stress, thereby enabling effective anti-tumor therapy. This study introduced a novel strategy to effectively promote the synergistic PTT/PDT effect by the enhanced oxidative stress.
Collapse
Affiliation(s)
- Yuanqi Yang
- Department of Microbial and Biochemical Pharmacy, School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Qingqing Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Wei Gu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Kaisheng Nan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Siyu Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Jinghai Zhang
- Department of Microbial and Biochemical Pharmacy, School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
4
|
Ma Q, Xu S, Wang Q, Que Y, He P, Yang R, Wang H, Wu Z, Xiao L, Yuan X, Li X, Xu T, Hu Y. Controllable All-in-One Biomimetic Hollow Nanoscaffold Initiating Pyroptosis-Mediated Antiosteosarcoma Targeted Therapy and Bone Defect Repair. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67424-67443. [PMID: 39603818 DOI: 10.1021/acsami.4c16287] [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: 11/29/2024]
Abstract
Pyroptosis has gained attention for its potential to reinvigorate the immune system within the tumor microenvironment. However, current approaches employing pyroptosis inducers suffer from limitations. They primarily rely on single agents, lack precise targeting, and potentially disrupt the intricate bone formation microenvironment, hindering local repair of tumor-induced bone defects. Therefore, a therapeutic strategy is urgently needed that can effectively trigger pyroptosis while simultaneously promoting bone regeneration. This research introduces an all-in-one construct designed to address these limitations. It combines a cell-camouflaged shell with an autosynergistic reactive oxygen species (ROS) generating polymer. This construct incorporates a hollow core of manganese dioxide (HMnO2) embedded with the photosensitizer IR780 and disguised by the cell membrane of an M1 macrophage. The M1 macrophage membrane grants the construct stealth-like properties, enabling it to accumulate selectively at the tumor site. Upon laser irradiation, IR780 acts as an exogenous trigger for ROS generation while simultaneously converting the light energy into heat. Additionally, the hollow structure of HMnO2 serves as an efficient carrier for IR780. Furthermore, Mn4+ ions released from HMnO2 deplete glutathione (GSH) within the tumor, further amplifying ROS production. This synergistic cascade ultimately culminates in pyroptosis induction through caspase-3-mediated cleavage of gasdermin E (GSDME) upon laser activation. Meanwhile, the depletion of GSH by HMnO2 within the tumor microenvironment (TME) leads to the generation of Mn2+ ions. These Mn2+ ions establish a supportive milieu, which promotes the transformation of bone marrow mesenchymal stem cells (BMSCs) into mature bone cells. This, in turn, promotes the repair of bone defects in rat femurs. Our findings strongly indicate that pyroptosis may be a strategy for osteosarcoma treatment, which presents a robust and versatile approach for targeted therapy and tissue regeneration in this patient population.
Collapse
Affiliation(s)
- Qiming Ma
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Shenglin Xu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Qian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Yukang Que
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Peng He
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Rui Yang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Hao Wang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Ziheng Wu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Longze Xiao
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Xingshi Yuan
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, Anhui 230001, China
| | - Xingxing Li
- Department of Orthopedics, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui 237008, China
| | - Tangbing Xu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Anhui Public Health Clinical Center, Hefei, Anhui 230012, China
| | - Yong Hu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| |
Collapse
|
5
|
Tagaras N, Song H, Sahar S, Tong W, Mao Z, Buerki‐Thurnherr T. Safety Landscape of Therapeutic Nanozymes and Future Research Directions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407816. [PMID: 39445544 PMCID: PMC11633477 DOI: 10.1002/advs.202407816] [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: 09/20/2024] [Indexed: 10/25/2024]
Abstract
Oxidative stress and inflammation are at the root of a multitude of diseases. Treatment of these conditions is often necessary but current standard therapies to fight excessive reactive oxygen species (ROS) and inflammation are often ineffective or complicated by substantial safety concerns. Nanozymes are emerging nanomaterials with intrinsic enzyme-like properties that hold great promise for effective cancer treatment, bacterial elimination, and anti-inflammatory/anti-oxidant therapy. While there is rapid progress in tailoring their catalytic activities as evidenced by the recent integration of single-atom catalysts (SACs) to create next-generation nanozymes with superior activity, selectivity, and stability, a better understanding and tuning of their safety profile is imperative for successful clinical translation. This review outlines the current applied safety assessment approaches and provides a comprehensive summary of the safety knowledge of therapeutic nanozymes. Overall, nanozymes so far show good in vitro and in vivo biocompatibility despite considerable differences in their composition and enzymatic activities. However, current safety investigations mostly cover a limited set of basic toxicological endpoints, which do not allow for a thorough and deep assessment. Ultimately, remaining research gaps that should be carefully addressed in future studies are highlighted, to optimize the safety profile of therapeutic nanozymes early in their pre-clinical development.
Collapse
Affiliation(s)
- Nikolaos Tagaras
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
- Department of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
| | - Haihan Song
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University866 Yuhangtang RdHangzhou310058China
| | - Shafaq Sahar
- College of Chemical and Biological EngineeringMOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University866 Yuhangtang RdHangzhou310058China
| | - Weijun Tong
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University866 Yuhangtang RdHangzhou310058China
| | - Zhengwei Mao
- College of Chemical and Biological EngineeringMOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University866 Yuhangtang RdHangzhou310058China
| | - Tina Buerki‐Thurnherr
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| |
Collapse
|
6
|
Yin H, Chen T, Hu X, Zhu W, Li Y, Sun W, Li L, Zhang H, Wang Q. Pyroptosis-Inducing Biomaterials Pave the Way for Transformative Antitumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2410336. [PMID: 39501932 DOI: 10.1002/advs.202410336] [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: 09/13/2024] [Revised: 10/15/2024] [Indexed: 12/19/2024]
Abstract
Pyroptosis can effectively overcome immunosuppression and reactivate antitumor immunity. However, pyroptosis initiation is challenging. First, the underlying biological mechanisms of pyroptosis are complex, and a variety of gasdermin family proteins can be targeted to induce pyroptosis. Second, other intracellular death pathways may also interfere with pyroptosis. The rationally designed gasdermin protein-targeting biomaterials are capable of inducing pyroptosis and have the capacity to stimulate antitumor immune function in a safe and effective manner. This review provides a comprehensive overview of the design, function, and antitumor efficacy of pyroptosis-inducing materials and the associated challenges, with a particular focus on the design options for pyroptosis-inducing biomaterials based on the activation of different gasdermin proteins. This review offers a valuable foundation for the further development of pyroptosis-inducing biomaterials for clinical applications.
Collapse
Affiliation(s)
- Hao Yin
- Institute for Advanced Research, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
- Department of Radiation and Medical Oncology, Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Tanzhou Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Xiaoqu Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Wenting Zhu
- Department of Oncology, Xijing Hospital of Air Force Military Medical University, Xi'an, Shaanxi, 710032, P. R. China
| | - Yida Li
- Department of Radiation and Medical Oncology, Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Wenjie Sun
- Department of Radiation and Medical Oncology, Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Lei Li
- The First Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, 710061, P. R. China
| | - Hongmei Zhang
- Department of Oncology, Xijing Hospital of Air Force Military Medical University, Xi'an, Shaanxi, 710032, P. R. China
| | - Qinyang Wang
- Institute for Advanced Research, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
- Department of Radiation and Medical Oncology, Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
- The First Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, 710061, P. R. China
| |
Collapse
|
7
|
Su Q, Wang Z, Zhou H, Zhang M, Deng W, Wei X, Xiao J, Duan X. Eradication of Large Tumors by Nanoscale Drug Self-Assembly. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410536. [PMID: 39420689 DOI: 10.1002/adma.202410536] [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: 07/20/2024] [Revised: 09/26/2024] [Indexed: 10/19/2024]
Abstract
Most patients with cancer are first diagnosed at an advanced disease stage, when tumors are already large and/or metastases are present. This circumstance has a negative impact on the prognosis and therapeutic effect of anticancer drugs. In this study, it is demonstrated that photosensitizer chlorin e6 and the photochemotherapy drug mitoxantrone self-assemble into relatively stable nanoassemblies (CM NAs) through hydrogen-bonding effect, π-π stacking, and hydrophobic interactions. Administration of CM NAs in combination with 660 nm laser irradiation shows chemotherapeutic, photothermal, and photodynamic effects, causing tumor cell apoptosis and pyroptosis and enabling noninvasive tumor ablation without compromising the surrounding normal tissue. More importantly, treatment with CM NAs increases tumor immunogenicity, leading to a strong and long-term antitumor immune response that eradicates large tumors and provides long-term protection against tumor recurrence on various tumor models.
Collapse
Affiliation(s)
- Qianyi Su
- Department of General Surgery, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhenyu Wang
- Department of General Surgery, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Cardiology, Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Huimin Zhou
- Department of General Surgery, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Miaomiao Zhang
- Department of General Surgery, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wenjia Deng
- Department of General Surgery, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xin Wei
- Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China
| | - Jisheng Xiao
- Department of General Surgery, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Cardiology, Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaopin Duan
- Department of General Surgery, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
8
|
Jia G, Wu Q, Hou M, Jiang Y, Yang H, Li M, Wu X, Zhang C. Ultrathin BSA-Stabilized Black Phosphorous Nanoreactor Boosts Mild-Temperature Photothermal Therapy Through Modulation of Cellular Self-Defense Fate. Adv Healthc Mater 2024; 13:e2402079. [PMID: 39225409 DOI: 10.1002/adhm.202402079] [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: 06/05/2024] [Revised: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Mild-temperature photothermal therapy (mild-PTT, 42-45 °C) offers a higher level of biosafety. However, its therapeutic effects are compromised by the heat shock response (HSR), a cellular self-defense mechanism, which triggers the overexpression of heat shock proteins (HSPs) with the capacity of repairing the damaged tumor cells. Herein, this work fabricates a novel nanoreactor by incorporating up-conversion nanoparticles (UCNPs), chlorin e6 (Ce6), and glucose oxidase (GOx) onto the ultrathin black phosphorus nanosheet (BPNS) (denoted as GOx-BUC). This nanoreactor amplifies mild-PTT effects under irradiation with an 808 nm laser, modulating HSPs-mediated cellular self-defense fate. On one hand, upon irradiation with a 980 nm laser, UCNPs can transfer energy to excite Ce6, leading to the generation of ROS burst, which achieves indiscriminate damage to HSPs activity in deeper tumor tissues. On the other hand, GOx can consume glucose, thereby depleting the ATP energy supply and further suppressing HSPs expression. Consequently, GOx-BUC exhibits excellent anti-tumor efficacy under mild temperature in a human colorectal cancer mouse model, resulting in complete tumor inhibition with negligible side effects. This black phosphorous nanoreactor, featuring dual-track HSPs destruction functionality, introduces novel perspectives for enhancing mild-PTT effectiveness while maintaining high biosafety.
Collapse
Affiliation(s)
- Guoping Jia
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qinghe Wu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Mengfei Hou
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yifei Jiang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Huizhen Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Meng Li
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xubo Wu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Chunfu Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| |
Collapse
|
9
|
Li X, Hu J, Zhao Q, Yao W, Jing Z, Jin Z. Towards precision medicine: design considerations for nanozymes in tumor treatment. J Transl Med 2024; 22:1033. [PMID: 39550581 PMCID: PMC11568558 DOI: 10.1186/s12967-024-05845-w] [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: 06/04/2024] [Accepted: 10/31/2024] [Indexed: 11/18/2024] Open
Abstract
Since the discovery of Fe3O4 nanoparticles with enzyme-like activity in 2007, nanozymes have emerged as a promising class of catalysts, offering advantages such as high catalytic efficiency, low cost, mild reaction conditions, and excellent stability. These properties make nanozymes highly suitable for large-scale production. In recent years, the convergence of nanomedicine and nanocatalysis has highlighted the potential of nanozymes in diagnostic and therapeutic applications, particularly in tumor therapy. Despite these advancements, the clinical translation of nanozymes remains hindered by the lack of designs tailored to specific tumor characteristics, limiting their effectiveness in targeted therapy. This review addresses the mechanisms by which nanozymes induce cell death in various tumor types and emphasizes the key design considerations needed to enhance their therapeutic potential. By identifying the challenges and opportunities in the field, this study aims to provide a foundation for future nanozyme development, ultimately contributing to more precise and effective cancer treatments.
Collapse
Affiliation(s)
- Xinqiao Li
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping district, Shenyang, 110001, People's Republic of China
| | - Jinpeng Hu
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping district, Shenyang, 110001, People's Republic of China
| | - Qi Zhao
- Department of Chemistry and the Institute for Sustainability and Energy, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208-3113, USA.
| | - Weifeng Yao
- Shanghai Key Laboratory of Materials Protection and Advanced Materials in Electric Power, College of Environmental & Chemical Engineering, Shanghai University of Electric Power, Shanghai, People's Republic of China.
- Shanghai Institute of Pollution Control and Ecological Security, Shanghai, People's Republic of China.
- Shanghai Engineering Research Center of Heat-Exchange System and Energy Saving, Shanghai University of Electric Power, Shanghai, People's Republic of China.
| | - Zhitao Jing
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping district, Shenyang, 110001, People's Republic of China.
| | - Zhizhong Jin
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping district, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
10
|
Zhu G, Xie Y, Wang J, Wang M, Qian Y, Sun Q, Dai Y, Li C. Multifunctional Copper-Phenolic Nanopills Achieve Comprehensive Polyamines Depletion to Provoke Enhanced Pyroptosis and Cuproptosis for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409066. [PMID: 39285820 DOI: 10.1002/adma.202409066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/08/2024] [Indexed: 11/08/2024]
Abstract
The overexpression of polyamines in tumor cells contributes to the establishment of immunosuppressive microenvironment and facilitates tumor growth. Here, it have ingeniously designed multifunctional copper-piceatannol/HA nanopills (Cu-Pic/HA NPs) that effectively cause total intracellular polyamines depletion by inhibiting polyamines synthesis, depleting intracellular polyamines, and impairing polyamines uptake, resulting in enhanced pyroptosis and cuproptosis, thus activating a powerful immune response to achieve anti-tumor therapy. Mitochondrial dysfunction resulting from overall intracellular polyamines depletion not only leads to the surge of copper ions in mitochondria, thereby causing the aggregation of toxic proteins to induce cuproptosis, but also triggers the accumulation of reactive oxygen species (ROS) within mitochondria, which further upregulates the expression of zDHHC5 and zDHHC9 to promote the palmitoylation of gasdermin D (GSDMD) and GSDMD-N, ultimately inducing enhanced pyroptosis. Then the occurrence of enhanced pyroptosis and cuproptosis is conductive to remodel the immunosuppressive tumor microenvironment, thus activating anti-tumor immune responses and ultimately effectively inhibiting tumor growth and metastasis. This therapeutic strategy of enhanced pyroptosis and cuproptosis through comprehensive polyamines depletion provides a novel template for cancer immunotherapy.
Collapse
Affiliation(s)
- Guoqing Zhu
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Yulin Xie
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Junrong Wang
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Man Wang
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Yanrong Qian
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Qianqian Sun
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Yunlu Dai
- Cancer Center and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, SAR, 999078, P. R. China
| | - Chunxia Li
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, P. R. China
| |
Collapse
|
11
|
Yang B, Cao L, Ge K, Lv C, Zhao Z, Zheng T, Gao S, Zhang J, Wang T, Jiang J, Qin Y. FeSA‐Ir/Metallene Nanozymes Induce Sequential Ferroptosis‐Pyroptosis for Multi‐Immunogenic Responses Against Lung Metastasis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401110. [PMID: 38874051 DOI: 10.1002/smll.202401110] [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: 02/12/2024] [Revised: 05/01/2024] [Indexed: 06/15/2024]
Abstract
For cancer metastasis inhibition, the combining of nanozymes with immune checkpoint blockade (ICB) therapy remains the major challenge in controllable reactive oxygen species (ROS) generation for creating effective immunogenicity. Herein, new nanozymes with light-controlled ROS production in terms of quantity and variety are developed by conjugating supramolecular-wrapped Fe single atom on iridium metallene with lattice-strained nanoislands (FeSA-Ir@PF NSs). The Fenton-like catalysis of FeSA-Ir@PF NSs effectively produced •OH radicals in dark, which induced ferroptosis and apoptosis of cancer cells. While under second near-infrared (NIR-II) light irradiation, FeSA-Ir@PF NSs showed ultrahigh photothermal conversion efficiency (𝜂, 75.29%), cooperative robust •OH generation, photocatalytic O2 and 1O2 generation, and caused significant pyroptosis of cancer cells. The controllable ROS generation, sequential cancer cells ferroptosis and pyroptosis, led 99.1% primary tumor inhibition and multi-immunogenic responses in vivo. Most importantly, the inhibition of cancer lung metastasis is completely achieved by FeSA-Ir@PF NSs with immune checkpoint inhibitors, as demonstrated in different mice lung metastasis models, including circulating tumor cells (CTCs) model. This work provided new inspiration for developing nanozymes for cancer treatments and metastasis inhibition.
Collapse
Affiliation(s)
- Baochan Yang
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
- School of Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 510260, China
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Lingzhi Cao
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, Hebei, 071002, China
| | - Kun Ge
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, Hebei, 071002, China
| | - Chaofan Lv
- School of Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 510260, China
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Zunling Zhao
- School of Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 510260, China
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Tianyu Zheng
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Shutao Gao
- College of Science, Hebei Agricultural University, Baoding, 071001, China
| | - Jinchao Zhang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, Hebei, 071002, China
| | - Tianyu Wang
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Jianzhuang Jiang
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Yan Qin
- School of Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 510260, China
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| |
Collapse
|
12
|
Xu X, Zhang Y, Meng C, Zheng W, Wang L, Zhao C, Luo F. Nanozymes in cancer immunotherapy: metabolic disruption and therapeutic synergy. J Mater Chem B 2024; 12:9111-9143. [PMID: 39177061 DOI: 10.1039/d4tb00769g] [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: 08/24/2024]
Abstract
Over the past decade, there has been a growing emphasis on investigating the role of immunotherapy in cancer treatment. However, it faces challenges such as limited efficacy, a diminished response rate, and serious adverse effects. Nanozymes, a subset of nanomaterials, demonstrate boundless potential in cancer catalytic therapy for their tunable activity, enhanced stability, and cost-effectiveness. By selectively targeting the metabolic vulnerabilities of tumors, they can effectively intensify the destruction of tumor cells and promote the release of antigenic substances, thereby eliciting immune clearance responses and impeding tumor progression. Combined with other therapies, they synergistically enhance the efficacy of immunotherapy. Hence, a large number of metabolism-regulating nanozymes with synergistic immunotherapeutic effects have been developed. This review summarizes recent advancements in cancer immunotherapy facilitated by nanozymes, focusing on engineering nanozymes to potentiate antitumor immune responses by disturbing tumor metabolism and performing synergistic treatment. The challenges and prospects in this field are outlined. We aim to provide guidance for nanozyme-mediated immunotherapy and pave the way for achieving durable tumor eradication.
Collapse
Affiliation(s)
- Xiangrui Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yaowen Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chijun Meng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenzhuo Zheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lingfeng Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chenyi Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Feng Luo
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu 610041, China.
| |
Collapse
|
13
|
Liu K, Zan P, Li Z, Lu H, Liu P, Zhang L, Wang H, Ma X, Chen F, Zhao J, Sun W. Engineering Bimetallic Polyphenol for Mild Photothermal Osteosarcoma Therapy and Immune Microenvironment Remodeling by Activating Pyroptosis and cGAS-STING Pathway. Adv Healthc Mater 2024; 13:e2400623. [PMID: 38691766 DOI: 10.1002/adhm.202400623] [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: 02/28/2024] [Revised: 04/28/2024] [Indexed: 05/03/2024]
Abstract
The immunosuppressive tumor microenvironment (ITME) of osteosarcoma (OS) poses a significant obstacle to the efficacy of existing immunotherapies. Despite the attempt of novel immune strategies such as immune checkpoint inhibitors and tumor vaccines, their effectiveness remains suboptimal due to the inherent difficulty in mitigating ITME simultaneously from both the tumor and immune system. The promotion of anti-tumor immunity through the induction of immunogenic cell death and activation of the cGAS-STING pathway has emerged as potential strategies to counter the ITME and stimulate systemic antitumor immune responses. Here, a bimetallic polyphenol-based nanoplatform (Mn/Fe-Gallate nanoparticles coated with tumor cell membranes is presented, MFG@TCM) which combines with mild photothermal therapy (PTT) for reversing ITME via simultaneously inducing pyroptosis in OS cells and activating the cGAS-STING pathway in dendritic cells (DCs). The immunostimulatory pathways, through the syngeneic effect, exerted a substantial positive impact on promoting the secretion of damage-associated molecular patterns (DAMPs) and proinflammatory cytokines, which favors remodeling the immune microenvironment. Consequently, effector T cells led to a notable antitumor immune response, effectively inhibiting the growth of both primary and distant tumors. This study proposes a new method for treating OS using mild PTT and immune mudulation, showing promise in overcoming current treatment limitations.
Collapse
Affiliation(s)
- Kaiyuan Liu
- Department of Bone Tumor Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P. R. China
| | - Pengfei Zan
- Department of Bone Tumor Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P. R. China
| | - Zihua Li
- School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Hengli Lu
- School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Peng Liu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, Guangdong, 518107, P. R. China
| | - Li Zhang
- School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Hongsheng Wang
- Department of Bone Tumor Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P. R. China
| | - Xiaojun Ma
- Department of Bone Tumor Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P. R. China
| | - Feng Chen
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, 200001, P. R. China
| | - Jing Zhao
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, Guangdong, 518107, P. R. China
| | - Wei Sun
- Department of Bone Tumor Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P. R. China
| |
Collapse
|
14
|
Cheng Q, Zhang T, Wang Q, Wu X, Li L, Lin R, Zhou Y, Qu S. Photocatalytic Carbon Dots-Triggered Pyroptosis for Whole Cancer Cell Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408685. [PMID: 39129656 DOI: 10.1002/adma.202408685] [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: 06/18/2024] [Revised: 07/25/2024] [Indexed: 08/13/2024]
Abstract
Manufacturing whole cancer cell vaccines (WCCV) with both biosafety and efficacy is crucial for tumor immunotherapy. Pyroptotic cancer cells, due to their highly immunogenic properties, present a promising avenue for the development of WCCV. However, the successful development of WCCV based on pyroptotic cancer cells is yet to be accomplished. Here, a facile strategy that utilized photocatalytic carbon dots (CDs) to induce pyroptosis of cancer cells for fabricating WCCV is reported. Photocatalytic CDs are capable of generating substantial amounts of hydroxyl radicals and can effectively decrease cytoplasmic pH values under white light irradiation. This process efficiently triggers cancer cell pyroptosis through the reactive oxygen species (ROS)-mitochondria-caspase 3-gasdermin E pathway and the proton motive force-driven mitochondrial ATP synthesis pathway. Moreover, in vitro, these photocatalytic CDs-induced pyroptotic cancer cells (PCIP) can hyperactivate macrophage (M0-M1) with upregulation of major histocompatibility complex class II expression. In vivo, PCIP induced specific immune-preventive effects in melanoma and breast cancer mouse models through anticancer immune memory, demonstrating effective WCCV. This work provides novel insights for inducing cancer cell pyroptosis and bridges the gap in the fabrication of WCCV based on pyroptotic cancer cells.
Collapse
Affiliation(s)
- Quansheng Cheng
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Tesen Zhang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Qingcheng Wang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Xue Wu
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Lingyun Li
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Runxing Lin
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Yinning Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
- Department of Physics and Chemistry, Faculty of Science and Technology, University of Macau, Taipa, Macau, SAR, 999078, China
- MOE Frontier Science Centre for Precision Oncology, Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Songnan Qu
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
- Department of Physics and Chemistry, Faculty of Science and Technology, University of Macau, Taipa, Macau, SAR, 999078, China
- MOE Frontier Science Centre for Precision Oncology, Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau, SAR, 999078, China
- Zhuhai UM Science and Technology Research Institute, Zhuhai, 519031, China
| |
Collapse
|
15
|
Li H, Chen Y, Gao Q, Wang N, Yang T, Du C, Chen M, Wang J. Modulating Visible-Light Driven NIR Lanthanide Polymer Photocatalysis for Amplification Detection of Exosomal Proteins and Cancer Diagnosis. Anal Chem 2024; 96:12084-12092. [PMID: 39001802 DOI: 10.1021/acs.analchem.4c02168] [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: 07/15/2024]
Abstract
Near-infrared (NIR) luminescent lanthanide materials hold great promise for bioanalysis, as they have anti-interference properties. The approach of efficient luminescence is sensitization through a reasonable chromophore to overcome the obstacle of the aqueous phase. The involvement of the surfactant motif is an innovative strategy to arrange the amphiphilic groups to be regularly distributed near the polymer to form a closed sensitized space. Herein, a lanthanide polymer (TCPP-PEI70K-FITC@Yb/SDBS) is designed in which the meso-tetra(4-carboxyphenyl)porphine (TCPP) ligand serves as both a sensitizer and photocatalytic switch. The surfactant sodium dodecyl benzenesulfonate (SDBS) wraps the photosensitive polymers to form a hydrophobic layer, which augments the light-harvesting ability and expedites its photocatalysis. TCPP-PEI70K-FITC@Yb/SDBS is subsequently applied as an amplified photocatalysis toolbox for universally regulating the generation of reactive oxygen species (ROS). Boosting 3,3',5,5'-tetramethylbenzidine (TMB) oxidation to produce blue products, a dual-mode biosensor is fabricated for improving the diagnosis of programmed death ligand-1-positive (PDL1) cancer exosomes. Exosomes were captured by Fe3O4 modified by the PDL1 aptamer, enabling replacement of alkaline phosphatase (ALP)-labeled multiple hybridized chains; then, the isolated ALP triggered a hydrolysis reaction to block the generation of oxTMB. Detection sensitivity improves by 1 order of magnitude through SDBS modulation, down to 104 particles/mL. The sensor performed well clinically in distinguishing cancer patients from healthy individuals, expanding physiological applications of near-infrared lanthanide luminescence.
Collapse
Affiliation(s)
- Haiyan Li
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Yafei Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Qing Gao
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Nan Wang
- Analytical and Testing Center, Northeastern University, Box 115, Shenyang 110819, China
| | - Ting Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Cheng Du
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Mingli Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
- Analytical and Testing Center, Northeastern University, Box 115, Shenyang 110819, China
| | - Jianhua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| |
Collapse
|
16
|
Liu P, Wu J, Chen L, Wu Z, Wu Y, Zhang G, Yu B, Zhang B, Wei N, Shi J, Zhang C, Lei L, Yu S, Lai J, Guo Z, Zheng Y, Jing Z, Jiang H, Wang T, Zhou J, Wu Y, Sun C, Shen J, Zhang J, Wu Z. Water-filtered infrared A radiation hyperthermia combined with immunotherapy for advanced gastrointestinal tumours. Cancer Med 2024; 13:e70024. [PMID: 39049187 PMCID: PMC11269209 DOI: 10.1002/cam4.70024] [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: 01/01/2024] [Revised: 06/18/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
This study pioneered the use of WIRA whole-body infrared hyperthermia combined with ICI therapy to treat GIT and verified the feasibility and safety of HIT. The final results showed a DCR of 55.6%, with a median PFS of 53.5 days, median OS of 134 days, and an irAE incidence of 22.2%. Therefore, we believe that HIT can exert multiple synergistic sensitisation effects, thereby providing clinical benefits to patients with advanced GITs, increasing overall safety, and improving patients' QOL.
Collapse
Affiliation(s)
- Pengyuan Liu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jing Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Liting Chen
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhenhai Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Yufei Wu
- ACS (International) School of SingaporeSingapore
| | - Ganlu Zhang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Bingqi Yu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Beibei Zhang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Nan Wei
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jinan Shi
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | | | - Lan Lei
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Shuhuan Yu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jianjun Lai
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhen Guo
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Yuli Zheng
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhao Jing
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Hao Jiang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | | | - Jueyi Zhou
- Department of OncologyLishui People's HospitalLishuiChina
| | - Yajun Wu
- TCM Dispensary, Zhejiang HospitalHangzhouChina
| | - Chuan Sun
- Geriatrics Institute of Zhejiang ProvinceDepartment of Geriatrics, Zhejiang HospitalHangzhouChina
| | - Jie Shen
- Department of Medical Oncology, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jian Zhang
- Department of Gastrointestinal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Zhibing Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
- Department of Radiation Oncology, Affiliated Zhejiang HospitalZhejiang University School of MedicineHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| |
Collapse
|
17
|
Jiang H, Huang X, Li H, Ren F, Li D, Liu Y, Tong Y, Ran P. Bacterial lipase-responsive polydopamine nanoparticles for detection and synergistic therapy of wound biofilms infection. Int J Biol Macromol 2024; 270:132350. [PMID: 38750839 DOI: 10.1016/j.ijbiomac.2024.132350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024]
Abstract
Wound biofilms represent an elusive conundrum in contemporary treatment and diagnostic options, accredited to their escalating antibiotic resistance and interference in chronic wound healing processes. Here, we developed mesoporous polydopamine (mPDA) nanoparticles, and grafted with rhodamine B (Rb) as biofilm lipase responsive detection probe, followed by π - π stacking mediated ciprofloxacin (CIP) loading to create mP-Rb@CIP nanoparticles. mPDA NPs with a melanin structure could quench fluorescence emissions of Rb. Once encountering biofilm in vivo, the ester bond in Rb and mPDA is hydrolyzed by elevated lipase concentrations, triggering the liberation of Rb and restore fluorescence emissions to achieve real-time imaging of biofilm-infected wounds. Afterwards, the 808 nm near-infrared (NIR) illumination initiates a spatiotemporal controlled antibacterial photothermal therapy (PTT), boosting its effectiveness through photothermal-triggered CIP release for synergistic biofilm eradication. The mP-Rb@CIP platform exhibits dual diagnostic and therapeutic functions, efficaciously treating biofilm-infected wounds in vivo and in vitro. Particularly, the mP-Rb@CIP/NIR procedure expedites wound-healing by alleviating oxidative stress, modulating inflammatory mediators, boosting collagen synthesis, and promoting angiogenesis. Taken together, the theranostic nanosystem strategy holds significant potential for addressing wound biofilm-associated infections.
Collapse
Affiliation(s)
- Hezhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Xiting Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Huanhuan Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Feifei Ren
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Dongqiu Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Yuan Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| | - Yan Tong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China.
| | - Pan Ran
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu Medical College, Chengdu 610051, China.
| |
Collapse
|
18
|
Jiang H, Fang W, Xu S, Luo H, Li D, Liu Y, Zeng Z, Tong Y, Zhao L. Synergistic quorum sensing inhibition and mild-temperature photothermal therapy of integrated nanoplatform for implant-associated biofilm infections. J Colloid Interface Sci 2024; 663:143-156. [PMID: 38401436 DOI: 10.1016/j.jcis.2024.02.155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/07/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
In current clinical practice, the presence of biofilms poses a significant challenge in the effective elimination of bacterial infections because of the physical and chemical barriers formed by biofilms, which offer persistent protection to bacteria. Here, we developed hollow mesoporous polydopamine (hMP) nanoparticles (NPs) loaded with luteolin (Lu) as a quorum sensing inhibitor, which were further coated with hyaluronic acid (HA) shells to create hMP-Lu@HA NPs. We observed that upon reaching the infection site, the HA shells underwent initial degradation by the hyaluronidase enzyme present in the bacterial infection's microenvironment to expose the hMP-Lu NPs. Subsequently, Lu was released in response to the acidic conditions characteristic of bacterial infections, which effectively hindered and dispersed the biofilm. Moreover, when subjected to near-infrared irradiation, the robust photothermal conversion effect of hMP NPs accelerated the release of Lu and disrupted the integrity of the biofilms by localized heating. This dual action enhanced the eradication of the biofilm infection. Importantly, hMP-Lu@HA NPs also promoted tissue regeneration and healing at the implantation site, concurrently addressing biofilm infection. Taken together, this nanosystem, combined with mild-temperature photothermal therapy and quorum sensing inhibition strategy, holds significant potential for applications in the treatment of implantation-associated infections.
Collapse
Affiliation(s)
- Hezhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Wenlan Fang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Shiqi Xu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Haimeng Luo
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Dongqiu Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Yuan Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhijun Zeng
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu Medical College, Chengdu 610051, China.
| | - Yan Tong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China.
| | - Long Zhao
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu Medical College, Chengdu 610051, China.
| |
Collapse
|
19
|
Chen P, Zhang C, He L, Li M, Rong J, Sun P, Chen Y, Li D. A thermoresponsive nanocomposite integrates NIR-II-absorbing small molecule with lonidamine for pyroptosis-promoted synergistic immunotherapy. J Nanobiotechnology 2024; 22:163. [PMID: 38600506 PMCID: PMC11007887 DOI: 10.1186/s12951-024-02424-5] [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: 02/15/2024] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Photothermal immunotherapy is regarded as the ideal cancer therapeutic modality to against malignant solid tumors; however, its therapeutic benefits are often modest and require improvement. In this study, a thermoresponsive nanoparticle (BTN@LND) composed of a photothermal agent (PTA) and pyroptosis inducer (lonidamine) were developed to enhance immunotherapy applications. Specifically, our "two-step" donor engineering strategy produced the strong NIR-II-absorbing organic small-molecule PTA (BTN) that exhibited high NIR-II photothermal performance (ε1064 = 1.51 × 104 M-1 cm-1, η = 75.8%), and this facilitates the diagnosis and treatment of deep tumor tissue. Moreover, the fabricated thermally responsive lipid nanoplatform based on BTN efficiently delivered lonidamine to the tumor site and achieved spatiotemporal release triggered by the NIR-II photothermal effect. In vitro and in vivo experiments demonstrated that the NIR-II photothermal therapy (PTT)-mediated on-demand release of cargo effectively faciliated tumor cell pyroptosis, thereby intensifying the immunogenic cell death (ICD) process to promote antitumor immunotherapy. As a result, this intelligent component bearing photothermal and chemotherapy can maximally suppress the growth of tumors, thus providing a promising approach for pyroptosis/NIR-II PTT synergistic therapy against tumors.
Collapse
Affiliation(s)
- Pengfei Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Liuliang He
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Mingfei Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jie Rong
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Pengfei Sun
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts & Telecommunications, Nanjing, 210023, China.
| | - Yingying Chen
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Daifeng Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
20
|
Wu X, Li Y, Wen M, Xie Y, Zeng K, Liu YN, Chen W, Zhao Y. Nanocatalysts for modulating antitumor immunity: fabrication, mechanisms and applications. Chem Soc Rev 2024; 53:2643-2692. [PMID: 38314836 DOI: 10.1039/d3cs00673e] [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: 02/07/2024]
Abstract
Immunotherapy harnesses the inherent immune system in the body to generate systemic antitumor immunity, offering a promising modality for defending against cancer. However, tumor immunosuppression and evasion seriously restrict the immune response rates in clinical settings. Catalytic nanomedicines can transform tumoral substances/metabolites into therapeutic products in situ, offering unique advantages in antitumor immunotherapy. Through catalytic reactions, both tumor eradication and immune regulation can be simultaneously achieved, favoring the development of systemic antitumor immunity. In recent years, with advancements in catalytic chemistry and nanotechnology, catalytic nanomedicines based on nanozymes, photocatalysts, sonocatalysts, Fenton catalysts, electrocatalysts, piezocatalysts, thermocatalysts and radiocatalysts have been rapidly developed with vast applications in cancer immunotherapy. This review provides an introduction to the fabrication of catalytic nanomedicines with an emphasis on their structures and engineering strategies. Furthermore, the catalytic substrates and state-of-the-art applications of nanocatalysts in cancer immunotherapy have also been outlined and discussed. The relationships between nanostructures and immune regulating performance of catalytic nanomedicines are highlighted to provide a deep understanding of their working mechanisms in the tumor microenvironment. Finally, the challenges and development trends are revealed, aiming to provide new insights for the future development of nanocatalysts in catalytic immunotherapy.
Collapse
Affiliation(s)
- Xianbo Wu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Yuqing Li
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Mei Wen
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Yongting Xie
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Ke Zeng
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - You-Nian Liu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Wansong Chen
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore.
| |
Collapse
|