1
|
Li M, Lu L, Guo Y, Fu J, Zhang Z, Li P, Guo Y, Han M, Wang X. Self-assembly of paclitaxel derivative and fructose as a potent inducer of immunogenic cell death to enhance cancer immunotherapy. Mater Today Bio 2025; 32:101793. [PMID: 40343162 PMCID: PMC12059333 DOI: 10.1016/j.mtbio.2025.101793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/27/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Immunotherapy shows promise for tumor control but is limited by low response rates. Paclitaxel (PTX) induces immunogenic cell death (ICD), yet conventional delivery systems face challenges like low drug loading and insufficient intracellular accumulation, reducing ICD efficacy. Small-molecule self-assembled PTX nanoparticles offer a promising solution due to high drug loading and dose delivery. In this study, PTX was conjugated with phenylboronic acid (PBA) to form the derivative PTX-PBA, which spontaneously self-assembled with fructose into nanoparticles (PTX-PBA-Fru NPs). These nanoparticles exhibited a uniform size of 107.8 ± 2.9 nm, a PDI of 0.064 ± 0.042, and a zeta potential of -12.2 ± 0.9 mV, with spherical morphology. In 4T1 tumor-bearing mice, PTX-PBA-Fru NPs significantly enhanced tumor inhibition (p < 0.001) and increased body weight (p < 0.05). No allergic reactions in healthy Balb/c mice and the maximum tolerated intravenous dose reached 200 mg/kg, underscoring its favorable safety profile of PTX-PBA-Fru NPs. The ICD effects induced by PTX-PBA-Fru NPs, when combined with the immunomodulator resiquimod (R848), elicited a robust anti-tumor immune response. This combination therapy effectively remodeled the immunosuppressive tumor microenvironment and achieved a 37.5 % tumor eradication rate. Moreover, it established long-term immune memory, providing protection against tumor re-challenge. This novel PTX formulation demonstrates strong anti-tumor effects, safety, and clinical potential in combination with R848-based immunotherapy.
Collapse
Affiliation(s)
- Manzhen Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing, 100193, China
| | - Likang Lu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing, 100193, China
| | - Yaoyao Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing, 100193, China
| | - Jingxin Fu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing, 100193, China
| | - Ziqi Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing, 100193, China
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, 150006, China
| | - Pengxin Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing, 100193, China
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yifei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing, 100193, China
| | - Meihua Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing, 100193, China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing, 100193, China
| |
Collapse
|
2
|
Dai Y, Lu S, Wei L, Liu L. Targeted Delivery of SmacN7 Peptide Induces Immunogenic Cell Death in Cervical Cancer Treatment. Appl Biochem Biotechnol 2025; 197:3295-3310. [PMID: 39862369 DOI: 10.1007/s12010-024-05129-5] [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] [Accepted: 11/20/2024] [Indexed: 01/27/2025]
Abstract
Cervical cancer is a common tumor in women and one of the common causes of cancer death in women. Due to the aggressive and non-specific nature of traditional chemotherapy, there is a growing need for new treatment modalities. Currently, tumor immunotherapy is increasingly garnering attention as a disruptive treatment approach. Therefore, we constructed CCTP-SmacN7, a delivery system capable of releasing active molecules in the tumor microenvironment. CCTP-SmacN7 can not only inhibit tumor proliferation and migration, but also induce tumors to produce large amounts of reactive oxygen species. The production of reactive oxygen species can activate tumors to release or expose damage-associated molecular patterns, promote DC cell maturation, and ultimately activate T cells. Here, we present an innovative targeted treatment approach for cervical cancer. While inducing tumor immunogenic cell death, this program can also improve the tumor microenvironment and initiate the tumor immune cycle.
Collapse
Affiliation(s)
- Yan Dai
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Shentao Lu
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Linna Wei
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Lubin Liu
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Wu X, Zhang J, Deng Z, Sun X, Zhang Y, Zhang C, Wang J, Yu X, Yang G. Bacteria-based biohybrids for remodeling adenosine-mediated immunosuppression to boost radiotherapy-triggered antitumor immune response. Biomaterials 2025; 316:123000. [PMID: 39674101 DOI: 10.1016/j.biomaterials.2024.123000] [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: 07/17/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
Radiotherapy (RT) can trigger immunogenic cell death (ICD) in tumor cells and release adenosine triphosphate (ATP) to activate antitumor immunity. However, the formation of immunosuppressive adenosine (ADO) mediated by ectonucleotidases including CD39 and CD73, can exacerbate the immunosuppressive effects. Herein, a radiosensitizer-based metal-organic framework (MOF) composed of bismuth (Bi) and ellagic acid (EA) was synthesized in situ on the surface of Escherichia coli Nissle 1917 (EcN) to serve as a carrier for the CD39 inhibitor sodium polyoxotungstate (POM-1). This therapeutic platform, acting as a radiosensitizer, significantly enhances cytotoxicity against tumor cells while effectively inducing ICD and releasing high concentrations of ATP. Subsequently, the released POM-1 increases the levels of pro-inflammatory extracellular ATP while preventing tumor immunosuppression caused by the accumulation of ADO. Additionally, as a natural immune adjuvant, EcN further promotes the maturation of dendritic cells (DCs) and the infiltration of cytotoxic T lymphocytes (CTLs). As a result, such treatment initiates the destruction of established tumor growth and induces strong abscopal effects, leading to a significant inhibition of tumor metastases. This strategy presents a bacterial-based biohybrid system that facilitates RT-induced ICD while simultaneously limiting the degradation of ATP into ADO, thereby achieving sustained anti-tumor immunity.
Collapse
Affiliation(s)
- Xirui Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Junjun Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou 215004, China
| | - Zheng Deng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xianglong Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yifan Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Cai Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jiadong Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xinke Yu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Guangbao Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| |
Collapse
|
4
|
Shao D, Pei X, Ma Y, Liu S, Li W, Li L, Ma P. Metallized hollow-COF nanobowls with dual-mode ROS generation for cancer sonodynamic therapy. J Mater Chem B 2025; 13:5181-5189. [PMID: 40223533 DOI: 10.1039/d5tb00338e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Sonodynamic therapy (SDT) has emerged as an encouraging route in tumor treatment, due to its exceptional tissue penetration depth and favorable safety profile. Nevertheless, the clinical translation of conventional organic sonosensitizers is hindered by intrinsic limitations, including pronounced hydrophobicity, insufficient chemical stability, and low reactive oxygen species (ROS) production. In contrast, hollow covalent organic frameworks (HCOFs) exhibit exceptional cargo-loading capabilities, structural robustness, and biocompatibility, positioning them as ideal nanoplatforms for advanced therapeutic applications. Herein, we engineered a bowl-shaped HCOF architecture designed to amplify ultrasonic cavitation effects. This nanostructure was subsequently functionalized with the sonosensitizer (Hemin) and subjected to strategic metallization via metal ion incorporation, culminating in the development of a high-efficiency antitumor nanosystem (FeHHCA). FeHHCA can achieve dual-mode ROS generation, namely, sonodynamic synergistically generating 1O2 and being specifically activated by a tumor microenvironment (TME) to generate ˙OH through a Fenton-like reaction, achieving an 78.7% tumor inhibition rate in vivo. These findings offer innovative approaches and strategies for the design of hollow COFs and offer great potential for the application of SDT in cancer treatment.
Collapse
Affiliation(s)
- Donghan Shao
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, 130022, China.
- Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528400, P. R. China
| | - Xinyu Pei
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, 130022, China.
- Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528400, P. R. China
| | - Yuqin Ma
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, 130022, China.
- Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528400, P. R. China
| | - Sainan Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China.
| | - Wenliang Li
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, 130022, China.
- Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528400, P. R. China
| | - Leijiao Li
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, 130022, China.
- Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528400, P. R. China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China.
- University of Science and Technology of China, Hefei 230026, P. R. China
| |
Collapse
|
5
|
Huang H, Tong QS, Zhang JY, Miao WM, Yu HH, Wang J, Shen S, Du JZ. Phagocytosis-Activating Nanocomplex Orchestrates Macrophage-Mediated Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2500982. [PMID: 40289887 DOI: 10.1002/adma.202500982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/09/2025] [Indexed: 04/30/2025]
Abstract
The phagocytosis of macrophages to tumor cells represents an alluring strategy for cancer immunotherapy; however, its effectiveness is largely hindered by the detrimental upregulation of anti-phagocytic signals and insufficient expression of pro-phagocytic signals of tumor cells. Here, a pro-phagocytic polymer-based nanocomplex is designed to promote the macrophage engulfment of tumor cells through concurrent modulation of both the "eat me" and "don't eat me" signals. The nanocomplex MNCCD47i-CALRt is formed by complexing a synthetic PAMAM derivative (G4P-C7A) that is capable of intrinsically inducing the exposure of calreticulin (CALR, a crucial pro-phagocytic protein) and a small inference RNA that can inhibit the expression of CD47 (a primary anti-phagocytic protein). MNCCD47i-CALRt can significantly delay tumor growth and prolong the survival of tumor-bearing mice with negligible hematopoietic toxicity in multiple murine colorectal cancer models. Furthermore, the pro-phagocytic capacity of MNCCD47i-CALRt is validated in the patient-derived tumor organoid model. Collectively, the phagocytosis-promoting nanocomplex provides a simple and potent strategy for boosting macrophage-mediated cancer immunotherapy.
Collapse
Affiliation(s)
- Hua Huang
- College & Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, 230032, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Qi-Song Tong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Jing-Yang Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Wei-Min Miao
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Hui-Han Yu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| |
Collapse
|
6
|
He S, Huang Y, Liu J, Liu H, Chen Y, Zou T, Sun J, Wang W, Wei H, Yu CY. A Metformin-Based Multifunctional Nanoplatform as a DNA Damage Amplifier for Maximized Radio-Immunotherapy to Overcome Radiotherapy Resistance. ACS NANO 2025; 19:14848-14864. [PMID: 40207668 DOI: 10.1021/acsnano.4c18627] [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: 04/11/2025]
Abstract
Radiotherapy (RT) has been highlighted to be an effective strategy for antitumor immunity activation by causing direct DNA damages, but it generally suffers from low response rates due to the compromised cytosolic DNA (cDNA) recognition by cyclic GMP-AMP synthase (cGAS). Simultaneous DNA repair and clearance system regulation for enhanced cDNA accumulation is a useful approach to improve immune response rates, which remains seldom reported to our knowledge. Here, we report the construction of a metformin (MET)-based multifunctional nanocomplex, CS-MET/siTREX1 (CSMT), consisting of biguanide-decorated CS (CS-MET) as the vector and 3'-5' DNA exonuclease TREX1 siRNA (siTREX1) as the therapeutic gene for RT-induced antitumor immunity enhancement by amplifying the initial DNA damage signals. The uniqueness of this study is the development of CSMT as a specific DNA damage amplifier to promote cDNA accumulation for maximizing radio-immunotherapy and circumventing RT resistance. Specifically, the CSMT nanocomplexes show not only enhanced gene transfection efficiency by MET modification but also synergistic therapeutic effects including MET's inhibition on DNA repair and siTREX1's attenuation on cDNA clearance, which leads to the greatest inhibitory effect in a Hepa1-6 proximal/distal tumor model with a high tumor growth inhibition (TGI) value of 99.1% for the primary tumor and significantly compromised distal tumor growth by inducing immunogenic cell death (ICD), promoting tumor-associated neutrophil (TAN) polarization, and stimulating tumor-specific memory T-cell generation. Overall, the CSMT nanocomplexes developed herein hold great translatable promises for overcoming RT resistance in clinics.
Collapse
Affiliation(s)
- Shuangyan He
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yun Huang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jia Liu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hongdu Liu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yalan Chen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Ting Zou
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jian Sun
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - WuZhou Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hua Wei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Cui-Yun Yu
- Affiliated Hospital of Hunan Academy of Chinese Medicine Hunan Academy of Chinese Medicine, Changsha 410013, China
| |
Collapse
|
7
|
Liu N, Wang X, Wang Z, Kan Y, Fang Y, Gao J, Kong X, Wang J. Nanomaterials-driven in situ vaccination: a novel frontier in tumor immunotherapy. J Hematol Oncol 2025; 18:45. [PMID: 40247328 PMCID: PMC12007348 DOI: 10.1186/s13045-025-01692-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
In situ vaccination (ISV) has emerged as a promising strategy in cancer immunotherapy, offering a targeted approach that uses the tumor microenvironment (TME) to stimulate an immune response directly at the tumor site. This method minimizes systemic exposure while maintaining therapeutic efficacy and enhancing safety. Recent advances in nanotechnology have enabled new approaches to ISV by utilizing nanomaterials with unique properties, including enhanced permeability, retention, and controlled drug release. ISV employing nanomaterials can induce immunogenic cell death and reverse the immunosuppressive and hypoxic TME, thereby converting a "cold" tumor into a "hot" tumor and facilitating a more robust immune response. This review examines the mechanisms through which nanomaterials-based ISV enhances anti-tumor immunity, summarizes clinical applications of these strategies, and evaluates its capacity to serve as a neoadjuvant therapy for eliminating micrometastases in early-stage cancer patients. Challenges associated with the clinical translation of nanomaterials-based ISV, including nanomaterial metabolism, optimization of treatment protocols, and integration with other therapies such as radiotherapy, chemotherapy, and photothermal therapy, are also discussed. Advances in nanotechnology and immunotherapy continue to expand the possible applications of ISV, potentially leading to improved outcomes across a broad range of cancer types.
Collapse
Affiliation(s)
- Naimeng Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yonemori Kan
- Department of Medical Oncology, National Cancer Center Hospital (NCCH), 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jidong Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518127, China.
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
8
|
Peng Y, Liu H, Liang X, Cao L, Teng M, Chen H, Li Z, Peng X, Mao J, Cheng H, Liu G. Self-assembling chemodrug fiber-hydrogel for transarterial chemoembolization and radiotherapy-enhanced antitumor immunity. J Control Release 2025; 380:1-16. [PMID: 39892652 DOI: 10.1016/j.jconrel.2025.01.088] [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: 12/17/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
Hydrogel, as a promising embolic material for hepatocellular carcinoma (HCC), may fully embolize both major vessels and peripheral microvessels. A self-assembling hydrogel composed of chemotherapeutic drugs offers significant clinical benefits without carrier introduction. Herein, we developed a sustained drug-releasing complex hydrogel (RKT@gel), which was fabricated by the self-assembly of raltitrexed chemotherapeutic drugs (R@gel), along with the incorporation of kaempferol and tantalum nanoparticles (Ta NPs). Kaempferol enhances the mechanical strength of R@gel and inhibits hypoxia-induced angiogenesis post-embolization, improving embolization effectiveness. In addition to enabling X-ray-guided transarterial chemoembolization (TACE), Ta NPs enhance radiation sensitivity. These synergistic effects of RKT@gel not only significantly induce immunogenic cell death, thereby enhancing the activation of dendritic cells, but also activate major histocompatibility complex class I (MHC-I)-mediated antitumor immune recognition and cytotoxicity. In vivo, RKT@gel achieves enhanced tumor deposition and sustained drug release, effectively suppressing tumor progression. Additionally, when combined with radiotherapy, RKT@gel achieves efficient antitumor immunoactivation. Overall, this versatile composite hydrogel not only achieves effective embolization therapy but also substantially triggers antitumor immune responses with good biocompatibility. This multifunctional design provides a TACE-based multidisciplinary strategy for HCC.
Collapse
Affiliation(s)
- Yisheng Peng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hui Liu
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiaoliu Liang
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Lei Cao
- Department of Pathology, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Minglei Teng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hu Chen
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhenjie Li
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xuqi Peng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jingsong Mao
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hongwei Cheng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China; Zhuhai UM Science & Technology Research Institute, University of Macau, Macau 999078, China.
| | - Gang Liu
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
9
|
Liu J, Liu J, Wang Y, Chen F, He Y, Xie X, Zhong Y, Yang C. Bioactive mesoporous silica materials-assisted cancer immunotherapy. Biomaterials 2025; 315:122919. [PMID: 39481339 DOI: 10.1016/j.biomaterials.2024.122919] [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: 07/14/2024] [Revised: 10/12/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
Immunotherapy is initially envisioned as a powerful approach to train immune cells within the tumor microenvironment (TME) and lymphoid tissues to elicit strong anti-tumor responses. However, clinical cancer immunotherapy still faces challenges, such as limited immunogenicity and insufficient immune response. Leveraging the advantages of mesoporous silica (MS) materials in controllable drug and immunomodulator release, recent efforts have focused on engineering MS with intrinsic immunoregulatory functions to promote robust, systemic, and safe anti-tumor responses. This review discusses advances in bioactive MS materials that address the challenges of immunotherapy. Beyond their role in on-demand delivery and drug release in response to the TME, we highlight the intrinsic functions of bioactive MS in orchestrating localized immune responses by inducing immunogenic cell death in tumor cells, modulating immune cell activity, and facilitating tumor-immune cell interactions. Additionally, we emphasize the advantages of bioactive MS in recruiting and activating immune cells within lymphoid tissues to initiate anti-tumor vaccination. The review also covers the challenges of MS-assisted immunotherapy, potential solutions, and future outlooks. With a deeper understanding of material-bio interactions, the rational design of MS with sophisticated bioactivities and controllable responsiveness holds great promise for enhancing the outcomes of personalized immunotherapy.
Collapse
Affiliation(s)
- Jiali Liu
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China
| | - Jiying Liu
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yaxin Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Yan He
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Xiaochun Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Yiling Zhong
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China.
| | - Chao Yang
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China.
| |
Collapse
|
10
|
Chu Z, Zheng W, Fu W, Liang J, Wang W, Xu L, Jiang X, Zha Z, Qian H. Implanted Microneedles Loaded with Sparfloxacin and Zinc-Manganese Sulfide Nanoparticles Activates Immunity for Postoperative Triple-Negative Breast Cancer to Prevent Recurrence and Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416270. [PMID: 40042034 PMCID: PMC12021102 DOI: 10.1002/advs.202416270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/15/2025] [Indexed: 04/26/2025]
Abstract
Recent investigations have underscored the significant role of manganese ions (Mn2+) in immunization, particularly through the activation of the cGAS-STING pathway, which enhances antitumor immune responses. However, the rapid metabolism of free Mn2+ following administration limits its effectiveness as an immune adjuvant. To address these challenges, microneedles (MNs) of hyaluronic acid containing Sparfloxacin (SP) and zinc-manganese sulfide (ZMS) are prepared for the postoperative in situ treatment of triple-negative breast cancer (TNBC) to prevent cancer recurrence and combat wound infection. ZMS/SP (ZS)-loaded MNs exhibit strong antimicrobial and antibiofilm properties that are crucial for preventing postoperative infections. Moreover, the generation of reactive oxygen species by these MNs disrupts the oxidative balance, effectively activating immunogenic cell death and facilitating the release of cytokines. ZS significantly suppressed tumor growth, reduced lung metastasis, and promoted wound healing. These effects are accompanied by notable increases in immune cell infiltration and activation, which is consistent with the gene sequencing results. Activation of the cGAS-STING pathway further improved antitumor immunity. These findings highlight the potential of ZS MNs as an effective and safe treatment that utilizes the immunostimulatory properties of Mn2+ to enhance local and systemic immune responses for the prevention of postoperative TNBC metastasis.
Collapse
Affiliation(s)
- Zhaoyou Chu
- School of Biomedical EngineeringAnhui Provincial Institute of Translational MedicineAnhui Medical UniversityHefeiAnhui230032P. R. China
- The First Affiliated Hospital of Anhui Medical UniversitHefeiAnhui230022P. R. China
| | - Wang Zheng
- School of Biomedical EngineeringAnhui Provincial Institute of Translational MedicineAnhui Medical UniversityHefeiAnhui230032P. R. China
| | - Wanyue Fu
- School of Biomedical EngineeringAnhui Provincial Institute of Translational MedicineAnhui Medical UniversityHefeiAnhui230032P. R. China
| | - Jun Liang
- School of Biomedical EngineeringAnhui Provincial Institute of Translational MedicineAnhui Medical UniversityHefeiAnhui230032P. R. China
| | - Wanni Wang
- School of Biomedical EngineeringAnhui Provincial Institute of Translational MedicineAnhui Medical UniversityHefeiAnhui230032P. R. China
| | - Lingling Xu
- School of Biomedical EngineeringAnhui Provincial Institute of Translational MedicineAnhui Medical UniversityHefeiAnhui230032P. R. China
| | - Xiaohua Jiang
- Department of Obstetrics and GynecologyCenter for Reproduction and GeneticsThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001P. R. China
| | - Zhengbao Zha
- School of Food and Biological EngineeringHefei University of TechnologyHefeiAnhui230009P. R. China
| | - Haisheng Qian
- School of Biomedical EngineeringAnhui Provincial Institute of Translational MedicineAnhui Medical UniversityHefeiAnhui230032P. R. China
| |
Collapse
|
11
|
He J, Ren X, Zhang Q, Wang S, Li Z, Cai K, Li M, Hu Y, Ran Q, Luo Z. Nanoradiosentizers with X ray-actuatable supramolecular aptamer building units for programmable immunostimulatory T cell engagement. Biomaterials 2025; 315:122924. [PMID: 39489019 DOI: 10.1016/j.biomaterials.2024.122924] [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: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
The insufficient activation and impaired effector functions of T cells in the immunosuppressive tumor microenvironment (TME) substantially reduces the immunostimulatory effects of radiotherapy. Herein, a multifunctional nanoradiosensitizer is established by integrating molecularly engineered aptamer precursors into cisplatin-loaded liposomes for enhancing radio-immunotherapy of solid tumors. Exposure to ionizing radiation (IR) following the nanoradiosensitizer treatment would induce pronounced immunogenic death (ICD) of tumor cells through cisplatin-mediated radiosensitization while also trigger the detachment of the aptamer precursors, which further self-assemble into PD-L1/PD-1-bispecific aptamer-based T cell engagers (CA) through the bridging effect of tumor-derived ATP to direct T cell binding onto tumor cells in the post-IR TME in a spatial-temporally programmable manner. The CA-mediated post-IR tumor-T cell engagement could override the immunosuppressive barriers in TME and enhance T cell-mediated recognition and elimination of tumor cells while minimizing systemic toxicities. Overall, this work offers an innovative approach to enhance the radio-immunotherapeutic efficacy in the clinics.
Collapse
Affiliation(s)
- Jinming He
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Xijiao Ren
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Qiqi Zhang
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Shuang Wang
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Zhongjun Li
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Qian Ran
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China.
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
12
|
Xu T, Liu K, Mi S, Yao Y, Zhang M, Xue S, Zhi F, Cryan SA, Ding D. Cyclooxygenase-2/prostaglandin E2 inhibition remodulated photodynamic therapy-associated immunosuppression for enhanced cancer immunotherapy. Mater Today Bio 2025; 31:101530. [PMID: 39990740 PMCID: PMC11847551 DOI: 10.1016/j.mtbio.2025.101530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/18/2025] [Accepted: 01/27/2025] [Indexed: 02/25/2025] Open
Abstract
Low immunogenicity and immunosuppressive tumor microenvironment (TME) are two pivotal factors restricting tumor immunotherapy. Photodynamic therapy (PDT) directly destroys cancer cells by producing reactive oxygen species (ROS), and enhances the immunogenicity of "cold" tumors by inducing immunogenic cell death (ICD), thereby promoting T cell development against tumors. However, PDT also deteriorates immunosuppression through overactivating the cyclooxygenase-2/prostaglandin E2 (COX-2/PGE2) pathway. To this end, biocompatible albumin nanoassemblies co-delivering IR780 and diclofenac are herein developed for enhanced therapy against triple-negative breast cancer. PDT-exacerbated PGE2 overexpression is effectively abolished by diclofenac-mediated COX-2 inhibition, which reprograms immunosuppressive TME via downregulating the infiltration of various immunosuppressive cells and their cytokine secretion to enhance effector T cell infiltration. Consequently, the enhanced antitumor immunity effectively inhibits tumor growth, prevents the recurrency and metastasis, and remarkably boosts the treatment efficacy of PD-L1 blockade. This study sets an intriguing example for overcoming the COX-2/PGE2 pathway-exacerbated immunosuppression alongside immune activation, thus enhancing synergistic cancer immunotherapy potentiated by various ROS-producing therapies (e.g., PDT and radiotherapy) and chemotherapy.
Collapse
Affiliation(s)
- Tao Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), Dublin, D02 YN77, Ireland
| | - Kehan Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Shuqi Mi
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yao Yao
- Department of Gerontology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, 223800, China
| | - Mengyao Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Shujuan Xue
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Feng Zhi
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, 213003, China
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Sally-Ann Cryan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), Dublin, D02 YN77, Ireland
| | - Dawei Ding
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China
- Jiangsu Province Higher Education Key Laboratory of Cell Therapy Nanoformulation (Construction), Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China
| |
Collapse
|
13
|
Liu D, Wang H, Yang W, Bai Y, Wu Z, Cui T, Bian K, Yi J, Shao C, Zhang B. One-Dose Bioorthogonal Gadolinium Nanoprobes for Prolonged Radiosensitization of Tumor. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2500504. [PMID: 40059485 DOI: 10.1002/smll.202500504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Indexed: 04/17/2025]
Abstract
Developing effective radiotherapy is impeded by tumor radioresistance, imprecise treatment, and the need for accurate imaging. Herein, a multifunctional gadolinium-based nanoprobe (GBD) is presented, integrating bioorthogonal click chemistry and theranostics to enhance tumor retention, magnetic resonance imaging (MRI) contrast, and radiosensitivity. GBD synthesis involved biomimetic mineralization of bovine serum albumin (BSA) with gadolinium ions to form nanoparticles (GB), followed by conjugation with dibenzocyclooctyne (DBCO). The optimized GBD exhibited an elevated longitudinal relaxivity (r1) of 25.54 mM-1 s-1, which represented a 6.7-fold enhancement compared to the clinical MRI contrast agent magnevist (Gd-DTPA, 3.81 mM-1 s-1). Notably, the application of bioorthogonal click chemistry enhanced the affinity and retention of GBD within tumor cells modified to express azide as an artificial receptor. This novel strategy enhanced tumor retention up to 16 days postinjection, outperforming DBCO-modified small molecule gadolinium (Gd-DBCO) with less than 1-day retention. Such prolonged retention facilitated continuous radiosensitization throughout the radiotherapy course, negating the need for multiple injections, and substantially boosted the effectiveness of radiotherapy. This study demonstrates the transformative potential of combining bioorthogonal click chemistry with nanotechnology in radiotherapy, offering a precise tumor targeting platform, real-time monitoring, and improved treatment outcomes.
Collapse
Affiliation(s)
- Dinghua Liu
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Hui Wang
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Weitao Yang
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Yang Bai
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhuoyao Wu
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Tianming Cui
- Shanghai Research Institute for Intelligent Autonomous Systems, Tongji University, Shanghai, 201210, China
| | - Kexin Bian
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jinyan Yi
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Bingbo Zhang
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200065, China
| |
Collapse
|
14
|
Zhu D, Lu M, Cheng H. NAT10 promotes radiotherapy resistance in non-small cell lung cancer by regulating KPNB1-mediated PD-L1 nuclear translocation. Open Life Sci 2025; 20:20251065. [PMID: 40109769 PMCID: PMC11920766 DOI: 10.1515/biol-2025-1065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 03/22/2025] Open
Abstract
Radiotherapy (RT) resistance in non-small cell lung cancer (NSCLC) is a significant contributor to tumor recurrence. NAT10, an enzyme that catalyzes ac4C RNA modification, has an unclear role in RT resistance. This study aimed to explore the function of NAT10 in RT resistance in NSCLC. RT-resistant NSCLC cell lines (PC9R and A549R) were established through repeated irradiation. The impact of NAT10 on cellular immunity was evaluated by measuring immune cell populations, cytotoxicity levels, and markers of cell dysfunction. Results demonstrated elevated levels of ac4C and NAT10 in RT-resistant cells. Knockdown of NAT10 suppressed cell proliferation and enhanced immune function in PC9R and A549R cells by upregulating TNF-α and IFN-γ while downregulating PD-1 and TIM-3. Mechanistically, RT resistance in NSCLC was mediated by NAT10-dependent ac4C modification of KPNB1. Furthermore, KPNB1 facilitated PD-L1 nuclear translocation, promoting immune escape in RT-resistant NSCLC cells. Overexpression of KPNB1 enhanced cell proliferation but impaired immune function in RT-resistant NSCLC cells. In conclusion, this study demonstrates that NAT10 upregulates KPNB1 expression through ac4C modification, thereby promoting RT resistance in NSCLC via PD-L1 nuclear translocation. These findings reveal a novel mechanism underlying RT resistance in NSCLC.
Collapse
Affiliation(s)
- Dagao Zhu
- Department of Radiation Oncology, The Affiliated Tongling Hospital of Bengbu Medical University, No. 468 Bijiashan Road, Tongguan District, Tongling, 244000, China
- Department of Radiation Oncology, The People's Hospital of Tongling City, No. 468 Bijiashan Road, Tongguan District, Tongling, 244000, China
| | - Mingliang Lu
- Department of Radiation Oncology, The Affiliated Tongling Hospital of Bengbu Medical University, No. 468 Bijiashan Road, Tongguan District, Tongling, 244000, China
| | - Hongmin Cheng
- Department of Radiation Oncology, The People's Hospital of Tongling City, No. 468 Bijiashan Road, Tongguan District, Tongling, 244000, China
| |
Collapse
|
15
|
Yang Y, Yang C, Deng K, Xiao Y, Liu X, Du Z. Nucleic Acid Drugs in Radiotherapy. Chembiochem 2025; 26:e202400854. [PMID: 39903093 DOI: 10.1002/cbic.202400854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/06/2025]
Abstract
Radiotherapy remains a cornerstone of cancer treatment, using high-energy radiation to induce DNA damage in tumor cells, leading to cell death. However, its efficacy is often hindered by challenges such as radiation resistance and side effects. As a powerful class of functional molecules, nucleic acid drugs (NADs) present a promising solution to these limitations. Engineered to target key pathways like DNA repair and tumor hypoxia, NADs can enhance radiotherapy sensitivity. NADs can also serve as delivery vehicles for radiotherapy agents such as radionuclides, improving targeting accuracy and minimizing side effects. This review explores the role of NADs in optimizing radiotherapy, highlighting their mechanisms, clinical applications, and synergies with radiotherapy, ultimately offering a promising strategy for improving patient outcomes in cancer therapy.
Collapse
Affiliation(s)
- Yuying Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Cai Yang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Kai Deng
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Yating Xiao
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, Universities and Colleges Admissions Service (UCAS), Hangzhou, 310024, China
| | - Xiangsheng Liu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Zhen Du
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
16
|
Yan Z, Bai Y, Zhang S, Kong L, Wang Y, Sun H, Li Y, Qiu L, Zhang R, Jiang P, Zhao D, Chen Z, Li Y, Pang H, Wang J. Quasi Fe MIL-53 nanozyme inducing ferroptosis and immunogenic cell death for cancer immunotherapy. Nat Commun 2025; 16:2290. [PMID: 40055308 PMCID: PMC11889140 DOI: 10.1038/s41467-025-57542-x] [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: 03/06/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
Nanozymes offer diverse therapeutic potentials for cancer treatment which is dependent on the development of nanomaterials. Quasi-metal-organic framework is a class of metal-organic framework-derived nanomaterials with a transition state from metal-organic frameworks towards metal oxide featuring porous structure and high activity. Herein an iron-based quasi-metal-organic framework nanozyme Q-MIL-53(Fe) is reported via a controlled deligandation strategy, exhibiting enhanced peroxidase-/catalase-mimic activity and glutathione depletion capacity, whose underlying mechanisms are studied via density functional theory calculations. Q-MIL-53(Fe) demonstrates biocompatibility and superior antitumor efficacy compared to pristine MIL-53(Fe). It can activate antitumor immune response by inducing ferroptosis and immunogenic cell death, promoting dendritic cell maturation and T lymphocytes infiltration. Furthermore, a combination of Q-MIL-53(Fe) and programmed cell death protein 1 antibody amplifies cancer immunotherapy. This study validates the antitumor activity of quasi-metal-organic frameworks and its immunotherapy induction potential. It would broaden the application of quasi-metal-organic frameworks and open avenues for developing antitumor nanozymes.
Collapse
Affiliation(s)
- Zihui Yan
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Yang Bai
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China.
| | - Songtao Zhang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225002, P. R. China
| | - Lingyi Kong
- Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Yu Wang
- Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Huilin Sun
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Yi Li
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Ruijie Zhang
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Pengju Jiang
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Donghui Zhao
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Zhongyan Chen
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, 325035, P. R. China
| | - Yafei Li
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China.
- Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China.
| | - Huan Pang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225002, P. R. China.
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China.
- School of Medical and Health Engineering, Changzhou University, Changzhou, 213164, P. R. China.
| |
Collapse
|
17
|
Jiang J, Lu Y, Zheng X, Xie M, Jauković A, Gao M, Zheng H. Engineering probiotic biohydrogen micro-factories to initiate reductive stress for boosting tumor vulnerability. Biomaterials 2025; 314:122892. [PMID: 39426122 DOI: 10.1016/j.biomaterials.2024.122892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/26/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
Disruption of redox homeostasis profoundly affects cellular metabolism and activities. While oxidative stress is extensively studied in cancer therapies, research on reductive stress remains in its infancy. Molecular hydrogen (H2), a well-known antioxidant, holds significant potential to induce reductive stress due to its strong antioxidative properties, making it a promising candidate for cancer therapy. However, it remains a major challenge to develop a sustainable H2 delivery system in vivo. Herein, we designed a micro-factory by engineering a gel-based microcapsule that encapsulates Enterobacter aerogenes, a.k.a. probiotic biohydrogen microcapsules (PBMCs), enabling the sustained H2 generation within tumor microenvironment. Notably, PBMCs effectively suppressed the proliferation of eight tumor cell lines as well as drug-resistant cancer cells. The prolonged H2 release from PBMCs induced reductive stress, as evidenced by a significant increase in the GSH/GSSG ratio in 4T1 cells. Moreover, PBMCs displayed significant antitumor effects in breast, melanoma and liver cancer models. The inhibition of PI3K-AKT pathway and the activation of MAPK pathway were identified as key mechanisms responsible for inducing tumor cell cycle arrest and apoptosis. The PBMCs also exhibited synergistic effects in combination with chemotherapeutics, resulting in robust inhibitions of preinvasive carcinoma growth and commonly associated pulmonary metastasis. Overall, our study introduces an innovative strategy to manipulate reductive stress in the tumor microenvironment through in situ H2 generation, thereby enhancing tumor vulnerability.
Collapse
Affiliation(s)
- Jie Jiang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Yuhao Lu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Xinyi Zheng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Maomao Xie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Aleksandra Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, PBOX 102, 11129, Belgrade, 11000, Serbia
| | - Meng Gao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China.
| | - Huizhen Zheng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
18
|
Guo Z, Huang T, Lv X, Yin R, Wan P, Li G, Zhang P, Xiao C, Chen X. Tumor microenvironment-activated polypeptide nanoparticles for oncolytic immunotherapy. Biomaterials 2025; 314:122870. [PMID: 39369669 DOI: 10.1016/j.biomaterials.2024.122870] [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: 07/31/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/08/2024]
Abstract
Cationic oncolytic polypeptides have gained increasing attention owing to their ability to directly lyse cancer cells and activate potent antitumor immunity. However, the low tumor cell selectivity and inherent toxicity induced by positive charges of oncolytic polypeptides hinder their systemic application. Herein, a tumor microenvironment-responsive nanoparticle (DNP) is developed by the self-assembly of a cationic oncolytic polypeptide (PLP) with a pH-sensitive anionic polypeptide via electrostatic interactions. After the formation of DNP, the positive charges of PLP are shielded. DNPs can keep stable in physiological conditions (pH 7.4) but respond to acidic tumor microenvironment (pH 6.8) to release oncolytic PLP. As a result, DNPs evoke potent immunogenic cell death by disrupting cell membranes, damaging mitochondria and increasing intracellular levels of reactive oxygen species. In vivo results indicate that DNPs significantly improve the biocompatibility of PLP, and inhibit tumor growth, recurrence and metastasis by direct oncolysis and activation of antitumor immune responses. In summary, these results indicate that pH-sensitive DNPs represent a prospective strategy to improve the tumor selectivity and biosafety of cationic polymers for oncolytic immunotherapy.
Collapse
Affiliation(s)
- Zhihui Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Tianze Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Xueli Lv
- College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Renyong Yin
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Pengqi Wan
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Gao Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China.
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| |
Collapse
|
19
|
Maiti D, Yu H, An JS, Yamashita S, Naito M, Miyata K, Kim HJ. Dual Porphyrin-Loaded Scintillating Nanoparticles Enhanced Photodynamic Therapy in Hypoxic Cancer Cells under X-ray Irradiation. Chembiochem 2025; 26:e202400838. [PMID: 39632271 DOI: 10.1002/cbic.202400838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 12/07/2024]
Abstract
Tumor hypoxia represents a major challenge to achieving successful therapy outcomes with photodynamic therapy (PDT). We hypothesized that systemic loading of dual porphyrins, protoporphyrin IX (PPIX) as a photosensitizer (PS) and hemin (Fe3+-PPIX) as an oxygen generator, onto Eu-doped NaYF4 scintillator (Sc), collectively terms as Eu-PPIX@Hemin, could enhance the activity of X-ray mediated PDT. Catalase-like property of hemin in the presence of H2O2 facilitated the production of oxygen molecules (3O2) in hypoxic cancer cells. The produced 3O2 reacts with nearby excited PPIX molecules (PPIX*) in the Sc-PS pairs to produce singlet oxygen (1O2), as cytotoxic reactive oxygen species (ROS) under X-ray irradiation. Eu-PPIX@Hemin nanoparticles (NPs) with a diameter of ~60 nm efficiently produced oxygen in the presence of H2O2, which its concentration in tumor cells is higher than that in normal cells. Eu-PPIX@Hemin generated similar amounts of ROS in hypoxic cultured cancer cells under low dose X-ray irradiation (0.5 Gy), compared to those in normoxic cancer cells. Notably, Eu-PPIX@Hemin exhibited similar cytotoxic effects in both hypoxic and normoxic cancer cells under X-ray irradiation. Overall, the mutual Sc-PS performance between PPIX and Eu was synergistically enhanced by hemin in Eu-PPIX@Hemin, which relieved hypoxia in the cancer cells under X-ray irradiation.
Collapse
Affiliation(s)
- Debabrata Maiti
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Hao Yu
- Nuclear Professional School, Graduate School of Engineering, The University of Tokyo, 2-22 Shirakata-shirane, Tokai-mura, Naka-gun, Ibaraki, 319-1188, Japan
| | - Jun Su An
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Shinichi Yamashita
- Nuclear Professional School, Graduate School of Engineering, The University of Tokyo, 2-22 Shirakata-shirane, Tokai-mura, Naka-gun, Ibaraki, 319-1188, Japan
| | - Mitsuru Naito
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Hyun Jin Kim
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
- Department of Biological Engineering, College of Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| |
Collapse
|
20
|
Li K, Yu X, Xu Y, Wang H, Liu Z, Wu C, Luo X, Xu J, Fang Y, Ju E, Lv S, Chan HF, Lao YH, He W, Tao Y, Li M. Cascaded immunotherapy with implantable dual-drug depots sequentially releasing STING agonists and apoptosis inducers. Nat Commun 2025; 16:1629. [PMID: 39952937 PMCID: PMC11828882 DOI: 10.1038/s41467-025-56407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/17/2025] [Indexed: 02/17/2025] Open
Abstract
Non-nucleotide stimulators of interferon gene (STING) agonists hold promise as immunotherapeutic agents for postsurgical adjuvant treatment of tumors. However, their limited effect duration hampers therapeutic effectiveness, necessitating prolonged administration of multiple doses that heightens infection risk and impacts patient compliance. Here, we develop an implantable dual-drug depot in a sandwich-like configuration, with a non-nucleotide STING agonist (MSA-2) in the outer layers of 3D-printed scaffolds and an immunogenic apoptosis inducer (doxorubicin, DOX) in the inner layer of electrospun fibers. We discover that MSA-2 can elicit endoplasmic reticulum stress-mediated and general immunogenic apoptosis of cancer cells. The stimulations with tumor-associated antigens and damage-associated molecular patterns from cancer cells, along with proinflammatory factors secreted by matured dendritic cells and M1-polarized macrophages, can depolymerize intracellular microtubules guiding activated STING trafficking towards lysosomes for degradation. Collectively, the dual-drug depots can initiate a long-lasting cascaded immunotherapy and chemotherapy, suppressing postsurgical tumor recurrence and metastasis.
Collapse
Affiliation(s)
- Kai Li
- Laboratory of Biomaterials and Translational Medicine, Department of Ultrasound, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Xuan Yu
- Laboratory of Biomaterials and Translational Medicine, Department of Ultrasound, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational Medicine, Department of Ultrasound, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Department of Ultrasound, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Zheng Liu
- Laboratory of Biomaterials and Translational Medicine, Department of Ultrasound, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Chong Wu
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xing Luo
- Laboratory of Biomaterials and Translational Medicine, Department of Ultrasound, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Jiancheng Xu
- Laboratory of Biomaterials and Translational Medicine, Department of Ultrasound, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Youqiang Fang
- Laboratory of Biomaterials and Translational Medicine, Department of Ultrasound, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Enguo Ju
- Laboratory of Biomaterials and Translational Medicine, Department of Ultrasound, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Weiling He
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Department of Ultrasound, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Department of Ultrasound, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China.
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-Sen University, Guangzhou, 510275, China.
| |
Collapse
|
21
|
Jia W, Wu Y, Xie Y, Yu M, Chen Y. Advanced Polymeric Nanoparticles for Cancer Immunotherapy: Materials Engineering, Immunotherapeutic Mechanism and Clinical Translation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413603. [PMID: 39797474 DOI: 10.1002/adma.202413603] [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: 09/10/2024] [Revised: 12/13/2024] [Indexed: 01/13/2025]
Abstract
Cancer immunotherapy, which leverages immune system components to treat malignancies, has emerged as a cornerstone of contemporary therapeutic strategies. Yet, critical concerns about the efficacy and safety of cancer immunotherapies remain formidable. Nanotechnology, especially polymeric nanoparticles (PNPs), offers unparalleled flexibility in manipulation-from the chemical composition and physical properties to the precision control of nanoassemblies. PNPs provide an optimal platform to amplify the potency and minimize systematic toxicity in a broad spectrum of immunotherapeutic modalities. In this comprehensive review, the basics of polymer chemistry, and state-of-the-art designs of PNPs from a physicochemical standpoint for cancer immunotherapy, encompassing therapeutic cancer vaccines, in situ vaccination, adoptive T-cell therapies, tumor-infiltrating immune cell-targeted therapies, therapeutic antibodies, and cytokine therapies are delineated. Each immunotherapy necessitates distinctively tailored design strategies in polymeric nanoplatforms. The extensive applications of PNPs, and investigation of their mechanisms of action for enhanced efficacy are particularly focused on. The safety profiles of PNPs and clinical research progress are discussed. Additionally, forthcoming developments and emergent trends of polymeric nano-immunotherapeutics poised to transform cancer treatment paradigms into clinics are explored.
Collapse
Affiliation(s)
- Wencong Jia
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Ye Wu
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
- Shanghai Institute of Materdicine, Shanghai, 200051, China
| |
Collapse
|
22
|
Cheng D, Luo L, Zhang Q, Song Z, Zhan Y, Tu W, Li J, Ma Q, Zeng X. Ca 2+- and cGAMP-Contained Semiconducting Polymer Nanomessengers for Radiodynamic-Activated Calcium Overload and Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411739. [PMID: 39679909 PMCID: PMC11809400 DOI: 10.1002/advs.202411739] [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: 09/23/2024] [Revised: 11/01/2024] [Indexed: 12/17/2024]
Abstract
Various second messengers exert some vital actions in biological systems, including cancer therapy, but the therapeutic efficacy is often need to be improved. A semiconducting polymer nanomessenger (TCa/SPN/a) consisting of two second messengers, calcium ion (Ca2+) and cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) for metastatic breast cancer therapy, is reported here. Such a TCa/SPN/a is constructed to exhibit X-ray response for the activatable delivery of mitochondria-targeting Ca compound and cGAMP as stimulator of interferon genes (STING) agonist. With X-ray irradiation, TCa/SPN/a could generate singlet oxygen (1O2) via radiodynamic effect for ablating solid tumors and improving the tumor immunogenicity by inducing immunogenic cell death (ICD). Furthermore, the released mitochondria-targeting Ca compounds show a high binging effect on mitochondria and cause reactive oxygen species (ROS) generation and mitochondria damage via calcium overload, while cGAMP boosts immunological effect through activating STING pathway. In this way, TCa/SPN/a enables a radiodynamic-activated calcium overload and immunotherapy to obviously inhibit the growths of bilateral tumors and also abolish tumor metastasis in metastatic breast cancer mouse models. This article should demonstrate the first smart dual-functional nanotherapeutic containing two second messengers for precise and specific cancer therapy.
Collapse
Affiliation(s)
- Danling Cheng
- Institute of ImmunologyZhejiang University School of MedicineHangzhou310009China
| | - Libai Luo
- Oncology Chemotherapy DepartmentAffiliated Hospital of Youjiang Medical University for Nationalities and Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education InstitutionsBaise533000China
| | - Qin Zhang
- Institute of Translational MedicineShanghai UniversityShanghai200444China
| | - Zheming Song
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Biological Science and Medical EngineeringDonghua UniversityShanghai201620China
| | - Yiduo Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Biological Science and Medical EngineeringDonghua UniversityShanghai201620China
| | - Wenzhi Tu
- Department of Radiation OncologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620China
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Biological Science and Medical EngineeringDonghua UniversityShanghai201620China
| | - Qiming Ma
- Department of General SurgeryThe First Affiliated Hospital of Gannan Medical UniversityGanzhou341000China
| | - Xianchang Zeng
- Institute of ImmunologyZhejiang University School of MedicineHangzhou310009China
| |
Collapse
|
23
|
Song Z, Hao Y, Long Y, Zhang P, Zeng R, Chen S, Chen W. Luminescent Lanthanide Infinite Coordination Polymers for Ratiometric Sensing Applications. Molecules 2025; 30:396. [PMID: 39860266 PMCID: PMC11767601 DOI: 10.3390/molecules30020396] [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: 12/31/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Ratiometric lanthanide coordination polymers (Ln-CPs) are advanced materials that combine the unique optical properties of lanthanide ions (e.g., Eu3+, Tb3+, Ce3+) with the structural flexibility and tunability of coordination polymers. These materials are widely used in biological and chemical sensing, environmental monitoring, and medical diagnostics due to their narrow-band emission, long fluorescence lifetimes, and excellent resistance to photobleaching. This review focuses on the composition, sensing mechanisms, and applications of ratiometric Ln-CPs. The ratiometric fluorescence mechanism relies on two distinct emission bands, which provides a self-calibrating, reliable, and precise method for detection. The relative intensity ratio between these bands varies with the concentration of the target analyte, enabling real-time monitoring and minimizing environmental interference. This ratiometric approach is particularly suitable for detecting trace analytes and for use in complex environments where factors like background noise, temperature fluctuations, and light intensity variations may affect the results. Finally, we outline future research directions for improving the design and synthesis of ratiometric Ln-CPs, such as incorporating long-lifetime reference luminescent molecules, exploring near-infrared emission systems, and developing up-conversion or two-photon luminescent materials. Progress in these areas could significantly broaden the scope of ratiometric Ln-CP applications, especially in biosensing, environmental monitoring, and other advanced fields.
Collapse
Affiliation(s)
- Ziqin Song
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China; (Z.S.); (Y.L.); (P.Z.); (R.Z.); (S.C.)
| | - Yuanqiang Hao
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China; (Z.S.); (Y.L.); (P.Z.); (R.Z.); (S.C.)
| | - Yunfei Long
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China; (Z.S.); (Y.L.); (P.Z.); (R.Z.); (S.C.)
| | - Peisheng Zhang
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China; (Z.S.); (Y.L.); (P.Z.); (R.Z.); (S.C.)
| | - Rongjin Zeng
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China; (Z.S.); (Y.L.); (P.Z.); (R.Z.); (S.C.)
| | - Shu Chen
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China; (Z.S.); (Y.L.); (P.Z.); (R.Z.); (S.C.)
| | - Wansong Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410017, China
| |
Collapse
|
24
|
Zhan S, Cao Z, Li J, Chen F, Lai X, Yang W, Teng Y, Li Z, Zhang W, Xie J. Iron Oxide Nanoparticles Induce Macrophage Secretion of ATP and HMGB1 to Enhance Irradiation-Led Immunogenic Cell Death. Bioconjug Chem 2025; 36:80-91. [PMID: 39680043 PMCID: PMC11740999 DOI: 10.1021/acs.bioconjchem.4c00488] [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: 10/26/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
ATP (adenosine triphosphate) and HMGB1 (high mobility group box 1 protein) are key players in treatments that induce immunogenic cell death (ICD). However, conventional therapies, including radiotherapy, are often insufficient to induce ICD. In this study, we explore a strategy using nanoparticle-loaded macrophages as a source of ATP and HMGB1 to complement radiation-induced intrinsic and adaptive immune responses. To this end, we tested three inorganic particles, namely, iron oxide nanoparticles (ION), aluminum oxide nanoparticles (AON), and zinc oxide nanoparticles (ZON), in vitro with bone marrow-derived dendritic cells (BMDCs) and then in vivo in syngeneic tumor models. Our results showed that ION was the most effective of the three nanoparticles in promoting the secretion of ATP and HMGB1 from macrophages without negatively affecting macrophage survival. Secretions from ION-loaded macrophages can activate BMDCs. Intratumoral injection of ION-loaded macrophages significantly enhanced tumor infiltration and activation of dendritic cells and cytotoxic T cells. Moreover, exogenous ION macrophages can enhance the efficacy of radiotherapy. In addition, direct injection of ION can also enhance the efficacy of radiotherapy, which is attributed to ION uptake by and stimulation of endogenous macrophages. Instead of directly targeting cancer cells, our strategy targets macrophages and uses them as a secretory source of ATP and HMGB1 to enhance radiation-induced ICD. Our research introduces a new nanoparticle-based immunomodulatory approach that may have applications in radiotherapy and beyond.
Collapse
Affiliation(s)
- Shuyue Zhan
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Zhengwei Cao
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Jianwen Li
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Fanghui Chen
- Department
of Hematology and Medical Oncology & Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Xinning Lai
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Wei Yang
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Yong Teng
- Department
of Hematology and Medical Oncology & Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Zibo Li
- Department
of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive
Cancer Center, University of North Carolina
at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Weizhong Zhang
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Jin Xie
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
25
|
Li Q, Wu Z, Chen S, Liang Y, Zhu K, Su N, Liu T, Zhao B. Enhancing Ferroptosis-Mediated Radiosensitization via Synergistic Disulfidptosis Induction. ACS NANO 2025; 19:1757-1770. [PMID: 39727204 DOI: 10.1021/acsnano.4c15932] [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: 12/28/2024]
Abstract
Ferroptosis plays an important role in radiotherapy (RT), and the induction of ferroptosis can effectively sensitize radiotherapy. However, the therapeutic efficiency is always affected by ferroptosis resistance, especially SLC7A11 (Solute Carrier Family 7 Member 11)-cystine-cysteine-GSH (glutathione)-GPX4 (glutathione peroxidase 4) pathway-mediated resistance. In this study, tumor-microenvironment self-activated high-Z element-containing nanoferroptosis inducers, PEGylated Fe-Bi-SS metal-organic frameworks (FBSP MOFs), were developed to sensitize RT. Unexpectedly, ferroptosis-resistant SLC7A11 would be self-adaptively upregulated, leading to self-responsive ferroptosis resistance. Since the conversion from SLC7A11-transported cystine to cysteine is highly glucose-dependent, glucose oxidase (GOx) was incorporated in the MOFs, causing the depletion of NADPH (nicotinamide adenine dinucleotide phosphate) to terminate the conversion from cystine to cysteine, relieving the self-adaptive ferroptosis resistance. Excitingly, the accumulation of cystine would synergistically lead to disulfide stress and trigger disulfidptosis, making a new contribution to enhance therapeutic efficiency. Moreover, the hydrogen peroxide produced during glucose oxidation could also cascade-react with the Fenton reaction, therefore enhancing ferropotosis. Both in vitro and in vivo results suggested that therapeutic efficiency of ferroptosis-mediated radiosensitization could be enhanced benefiting from synergistic disulfidptosis induction, indicating that disulfidptosis might be an efficient strategy to relieve ferroptosis resistance and enhance RT efficiency.
Collapse
Affiliation(s)
- Qiuyu Li
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
| | - Zede Wu
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P.R. China
| | - Siwen Chen
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
| | - Yu Liang
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kai Zhu
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P.R. China
| | - Ning Su
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tiancai Liu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, P.R. China
| | - Bingxia Zhao
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
- Experiment Education/Administration Center, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, P.R. China
| |
Collapse
|
26
|
Li W, Zhang S, Liu L, Li M, He J, Meng Q, Kang J, Zhou D, Gao L, Bai J, Gu Z, Gao F. Enhancing Chordoma Radiotherapy: Ta@PVP Nanoparticles as Potent Radiosensitizers. ACS APPLIED MATERIALS & INTERFACES 2025; 17:750-762. [PMID: 39693110 DOI: 10.1021/acsami.4c19601] [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: 12/19/2024]
Abstract
Surgical resection and high-dose radiotherapy constitute the standard therapeutic approaches for chordoma. However, the efficacy of radiotherapy is often compromised by the tumor microenvironment's hypoxic conditions, which confer radiation resistance, and by the potential damage to adjacent spinal cord and neural structures from elevated radiation doses. To address these challenges, we employed high biocompatible poly(vinylpyrrolidone)-modified tantalum nanoparticles (Ta@PVP NPs) as a potent radiosensitizer to augment the radiotherapy sensitivity of chordoma. Upon exposure to X-ray irradiation, Ta@PVP NPs demonstrated the capability to efficiently deposit X-ray radiation energy within the tumor microenvironment, subsequently generating reactive oxygen species (ROS) that induce oxidative stress in the tumor. Both in vitro and in vivo experiments revealed that Ta@PVP NPs significantly enhanced the cytotoxic effects of X-ray, thereby markedly inhibiting the proliferation of chordoma cells and impeding tumor growth. This study explored the radiosensitization potential of Ta@PVP NPs in the context of chordoma, highlighting the application of radiosensitizers as a promising strategy to augment the efficacy of chordoma radiotherapy.
Collapse
Affiliation(s)
- Wancheng Li
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Department of Neurosurgery, Anshan Central Hospital of China Medical University, Anshan 114000, China
| | - Shuheng Zhang
- Department of Neurosurgery, Anshan Central Hospital of China Medical University, Anshan 114000, China
| | - Linhong Liu
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Mingxuan Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Jinfeng He
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Qingguo Meng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Jiali Kang
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Dabiao Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Liang Gao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Jiwei Bai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Zhanjun Gu
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Fuping Gao
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
- Spallation Neutron Source Science Center, Dongguan 523803, China
- Jinan Laboratory of Applied Nuclear Science, Jinan 251401, China
| |
Collapse
|
27
|
Wang M, Zhao B, Huang A, Song M, Wang J, Liu P. Radiation Therapy Improves Survival in Patients with Distant Metastatic Head and Neck Squamous Cell Carcinoma: A Retrospective Study. J Cancer 2025; 16:996-1007. [PMID: 39781346 PMCID: PMC11705048 DOI: 10.7150/jca.105254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND The role of radiation therapy in patients with distant metastatic squamous cell carcinoma of the head and neck (mHNSCC) is unclear. In this study, we compare the differences in survival among mHNSCC patients treated with chemotherapy plus radiotherapy (RT) vs. chemotherapy alone. MATERIALS AND METHODS This study included patients with distant mHNSCC recruited from 2 cohorts: The Surveillance, Epidemiology, and End Results (SEER [N=885]) database and a Chinese single-institution registry in Sun Yat-sen University Cancer Center (SYSUCC [N=60]). The SEER cohort included 600 patients received RT plus chemotherapy and 285 patients received chemotherapy alone; in the SYSUCC cohort, 40 patients received RT plus chemotherapy and 20 patients received chemotherapy alone were recruited. The period of data collection for the SEER study was from January 2010 to December 2015, and that for SYSUCC was from January 2010 to December 2020. The study's primary outcome was overall survival (OS), with disease-specific survival (DSS) as a secondary outcome. RESULTS Of the 885 patients in the SEER cohort, the addition of RT to chemotherapy increased one-year OS from 46.6% to 56.0% compared with chemotherapy alone (P =0.009) and from 10.4% to 25.1% for three-year OS (P <0.001). Patients who received RT in addition to chemotherapy were also more likely to have better three-year DSS than those who received chemotherapy alone (28.9% vs. 14.0%, P<0.001). Similarly, in the SYSUCC cohort, patients who received chemotherapy plus RT had better three-year OS than chemotherapy alone (62.8% vs. 21.0%, P=0.003). The addition of RT to chemotherapy increased median OS among patients with mHNSCC from 10 months to 13 months in the SEER cohort and from 14 months to 29 months in the SYSUCC cohort. CONCLUSION Radiotherapy in addition to chemotherapy significantly improved OS and DSS in patients with mHNSCC.
Collapse
Affiliation(s)
- Mengwen Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Baitian Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Department of Clinical Trials Center, China
| | - Amin Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Department of medical oncology, the First Affiliated Hospital, Sun Yat-sen University, China
| | - Ming Song
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jia Wang
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
| | - Panpan Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
28
|
Zheng R, Yu C, Yao D, Cai M, Zhang L, Ye F, Huang X. Engineering Stimuli-Responsive Materials for Precision Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406439. [PMID: 39444066 PMCID: PMC11707583 DOI: 10.1002/smll.202406439] [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: 07/28/2024] [Revised: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Over the past decade, precision medicine has garnered increasing attention, making significant strides in discovering new therapeutic drugs and mechanisms, resulting in notable achievements in symptom alleviation, pain reduction, and extended survival rates. However, the limited target specificity of primary drugs and inter-individual differences have often necessitated high-dosage strategies, leading to challenges such as restricted deep tissue penetration rates and systemic side effects. Material science advancements present a promising avenue for these issues. By leveraging the distinct internal features of diseased regions and the application of specific external stimuli, responsive materials can be tailored to achieve targeted delivery, controllable release, and specific biochemical reactions. This review aims to highlight the latest advancements in stimuli-responsive materials and their potential in precision medicine. Initially, we introduce disease-related internal stimuli and capable external stimuli, elucidating the reaction principles of responsive functional groups. Subsequently, we provide a detailed analysis of representative pre-clinical achievements of stimuli responsive materials across various clinical applications, including enhancements in the treatment of cancers, injury diseases, inflammatory diseases, infection diseases, and high-throughput microfluidic biosensors. Finally, we discuss some clinical challenges, such as off-target effects, long-term impacts of nano-materials, potential ethical concerns, and offer insights into future perspectives of stimuli-responsive materials.
Collapse
Affiliation(s)
- Ruixuan Zheng
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Chang Yu
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
- Intervention DepartmentThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Dan Yao
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Mengsi Cai
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Lexiang Zhang
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325000China
| | - Fangfu Ye
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325000China
- Beijing National Laboratory for Condensed Matter PhysicsInstitute of PhysicsChinese Academy of SciencesBeijing100190China
| | - Xiaoying Huang
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| |
Collapse
|
29
|
Sun S, Hou X, Li K, Huang C, Rong Y, Bi J, Li X, Wu D. Curcumin and Metformin Infinite Coordination Polymer Nanoparticles for Combined Therapy of Diabetic Mice via Intraperitoneal Injections. J Funct Biomater 2024; 15:388. [PMID: 39728188 DOI: 10.3390/jfb15120388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/01/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024] Open
Abstract
Metformin (Met) is one of the most commonly prescribed first-line drugs for diabetes treatment. However, it has several issues, including low bioavailability, therapeutic platform, and side effects at high doses. In order to improve the therapeutic efficiency of Met, this study proposes a strategy of using Met and curcumin (Cur) to prepare Cur-Zn(II)-Met infinite coordination polymer nanoparticles (CM ICP NPs), and combining this with intraperitoneal injections, for the treatment of diabetic mice. Fourier transform infrared (FTIR) spectroscopy, X-ray photoelectron spectroscopy (XPS), transmission electron microscopy (TEM), nanoparticle analysis, cytotoxicity experiments, and mice experiments were used to investigate structure, properties, and application effects. The results showed that CM ICP NPs exhibit a high drug encapsulation rate (100%), good stability, and an absence of in vivo and in vitro toxicity. The blood glucose level of diabetic mice after treatment was reduced to 6.7 ± 0.65 mmol/L at the seventh week. In terms of therapeutic mechanism, it appears that Met and Cur can synergistically regulate blood glucose in mice from multiple paths. This study provides a promising method for the treatment of diabetes using Met and other drugs.
Collapse
Affiliation(s)
- Siwei Sun
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
| | - Xinyi Hou
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
- School of Pharmacy, Xi'an Medical University, Xi'an 710021, China
| | - Ke Li
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
| | - Chenqi Huang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yu Rong
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
| | - Jiao Bi
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
| | - Xueping Li
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
- School of Clinical Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
30
|
Xu M, Xu C, Qiu Y, Feng Y, Shi Q, Liu Y, Deng H, Ma X, Lin N, Shi Q, Shen Z, Meng S, Yang J, Chen H, Xue F. Zinc-based radioenhancers to activate tumor radioimmunotherapy by PD-L1 and cGAS-STING pathway. J Nanobiotechnology 2024; 22:782. [PMID: 39702231 DOI: 10.1186/s12951-024-02999-z] [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: 04/18/2024] [Accepted: 11/06/2024] [Indexed: 12/21/2024] Open
Abstract
Radiotherapy and immunotherapy have already become the primary form of treatment for non-small-cell lung cancer (NSCLC), but are limited by high radiotherapy dose and low immune response rate. Herein, a multi-pronged strategy using a radio-immuno-enhancer (ZnO-Au@mSiO2) is developed by inducing tumor cells apoptosis and reprograming the immunosuppressive tumor microenvironment (TME). The radio-immuno-enhancer employed Au as a radiosensitizer, transition Zn ions as immune activators, which not only tremendously enhances the anti-proliferative activity of radiotherapy toward cancer cells, but also activates the immune response with multi-targets to let "exhausted" T cells "back to life" by triggering immunogenic cell death (ICD), immune checkpoint blockade (ICB) that target PD-1/PD-L1 and cGAS-STING under X-ray irradiation with a low dosage. The in vivo results demonstrate desirable antitumor and immunogenic effects of radio-immuno-enhancer-mediated immune activation by increasing the ratio of cytotoxic T cells (CTLs) and helper T cells. This work provides a feasible approach for future development of effective transition metal ion-activated radio-immunotherapeutic agents.
Collapse
Affiliation(s)
- Mengjiao Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Chao Xu
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Yu Qiu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China
| | - Yushuo Feng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China
| | - Qianqian Shi
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China
| | - Yaqing Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China
| | - Huaping Deng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China
| | - Xiaoqian Ma
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China
| | - Nuo Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China
| | - Qunying Shi
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China
| | - Zhiyang Shen
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China
| | - Shanshan Meng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China
| | - Jiang Yang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hongmin Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, Xiamen, China.
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China.
| | - Fangqin Xue
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China.
| |
Collapse
|
31
|
Sun Y, Chen Y, Wu B, Li H, Wang Y, Wang X, Deng L, Yang K, Wang X, Cheng W. Synergistic SDT/cuproptosis therapy for liver hepatocellular carcinoma: enhanced antitumor efficacy and specific mechanisms. J Nanobiotechnology 2024; 22:762. [PMID: 39696275 DOI: 10.1186/s12951-024-02995-3] [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: 09/04/2024] [Accepted: 11/06/2024] [Indexed: 12/20/2024] Open
Abstract
The efficacy of sonodynamic therapy (SDT), an emerging approach for tumor treatment, is hindered by the high levels of the antioxidant glutathione (GSH) in the tumor microenvironment (TME). In this study, we constructed nanobubbles loaded with the sonosensitizer HMME and the tumor-targeting peptide RGD (HMME-RGD@C3F8 NBs) for synergistic SDT/cuproptosis therapy of liver hepatocellular carcinoma (LIHC) in combination with Elesclomol-Cu as cuproptosis inducers. Endogenous GSH is consumed by Cu2+ to modulate the complex TME, thereby amplifying oxidative stress and further improving SDT performance. Additionally, intracellular Cu2+ overload can induce cuproptosis, which is further amplified by SDT, to initiate irreversible protein toxicity. The specific mechanism of synergistic SDT/cuproptosis therapy in LIHC was investigated by RNA sequencing analysis. The synergistic SDT/cuproptosis therapy reprogrammed the TME to improve the efficacy of immune checkpoint inhibitor-based immunotherapy. Furthermore, a risk-scoring model was created and displayed significant promise in the prognosis of LIHC.
Collapse
Affiliation(s)
- Yucao Sun
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Yichi Chen
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Bolin Wu
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Helin Li
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Yijun Wang
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Xiaodong Wang
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Liwen Deng
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Kuikun Yang
- School of Life Science and Technology, Harbin Institute of Technology, No. 92, Xidazhi Street, Nangang District, Harbin, 150081, Heilongjiang, China.
| | - Xiuhong Wang
- Department of Biochemistry and Molecular Biology, Heilongjiang Provincial Science and Technology Innovation Team in Higher Education Institutes for Infection and Immunity, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China.
| | - Wen Cheng
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
32
|
Cao X, Feng N, Huang Q, Liu Y. Nanoscale Metal-Organic Frameworks and Nanoscale Coordination Polymers: From Synthesis to Cancer Therapy and Biomedical Imaging. ACS APPLIED BIO MATERIALS 2024; 7:7965-7986. [PMID: 38382060 DOI: 10.1021/acsabm.3c01300] [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: 02/23/2024]
Abstract
Recently, there has been significant interest in nanoscale metal-organic frameworks (NMOFs) characterized by ordered crystal structures and nanoscale coordination polymers (NCPs) featuring amorphous structures. These structures arise from the coordination interactions between inorganic metal ions or clusters and organic ligands. Their advantages, such as the ability to tailor composition and structure, efficiently encapsulate diverse therapeutic or imaging agents within porous frameworks, inherent biodegradability, and surface functionalization capability, position them as promising carriers in the biomedical fields. This review provides an overview of the synthesis and surface modification strategies employed for NMOFs and NCPs, along with their applications in cancer treatment and biological imaging. Finally, future directions and challenges associated with the utilization of NMOFs and NCPs in cancer treatment and diagnosis are also discussed.
Collapse
Affiliation(s)
- Xianghui Cao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, China
| | - Nana Feng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, China
| | - Qingqing Huang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, China
| | - Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, China
| |
Collapse
|
33
|
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
|
34
|
Liao Y, Wang D, Gu C, Wang X, Zhu S, Zheng Z, Zhang F, Yan J, Gu Z. A cuproptosis nanocapsule for cancer radiotherapy. NATURE NANOTECHNOLOGY 2024; 19:1892-1902. [PMID: 39300223 DOI: 10.1038/s41565-024-01784-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
Residual tumours that persist after radiotherapy often develop acquired radiation resistance, increasing the risk of recurrence and metastasis while providing obstacles to re-irradiation. Using samples from patients and experimental mice, we discovered that FDX1 and LIAS, key regulators of cuproptosis, were up-regulated in residual tumours following radiotherapy, conferring the increased sensitivity to cuproptosis. Therefore, we proposed a novel radiosensitization strategy focused on cuproptosis, using a copper-containing nanocapsule-like polyoxometalate as a paradigm. In an initial demonstration, we showed that the nanocapsule released copper ions in a controlled manner upon exposure to ionizing radiation. Furthermore, radiation-triggered cuproptosis overcame acquired radiation resistance even at clinically relevant radiation doses and activated a robust abscopal effect, with a 40% cure rate in both radioresistant and re-irradiation tumour models. Collectively, targeting cuproptosis is a compelling strategy for addressing acquired radiation resistance, optimizing the local antitumour effects of radiotherapy while simultaneously activating systemic antitumour immunity.
Collapse
Affiliation(s)
- You Liao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Dongmei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Chenglu Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Xue Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Ziye Zheng
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fuquan Zhang
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junfang Yan
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China.
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
35
|
Lin F, Qin Y, Sun J, Liu Y, Yang S, Zheng S, Yin L, Li D, Cui L, Li G, Qiu Z, Liu Z. Delivery of Cu(II) and Mn(II) by polydopamine-modified nanoparticles for combined photothermal and chemotherapy. J Mater Chem B 2024; 12:12062-12072. [PMID: 39445457 DOI: 10.1039/d4tb01819b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Chemodynamic therapy (CDT) has been recognized as an emerging therapeutic strategy. It has attracted considerable attention in recent years as it can generate the most harmful reactive oxygen species (ROS)-hydroxyl radicals (•OH) through the Fenton reaction or a Fenton-like reaction under the catalysis of versatile metal cations, such as, Fe(II), Fe(III), Cu(I), Mn(II), and Mn(III). However, a large number of reducing species (e.g., GSH) in tumors inhibit the therapeutic effects of CDT. This study proposes a nanocarrier strategy that can release versatile metal cations in the initial stage to consume the reducing substances, which can be convenient for subsequent CDT treatment. A novel nano-delivery system based on H-MnO2@PDA/Cu-CD@Ad-TK-Ad@Ploy-CD (abbreviated as MNZ) was proposed to resolve the above problems. Herein, hollow mesoporous manganese dioxide nanoparticles (H-MnO2) were coated with PDA and modified with copper ions on the surface of PDA. The PDA was then functionalized with β-cyclodextrin (β-CD) substitutions that were further assembled with N-((1S,3R,5S)-adamantan-1-yl)-3-((2-((3-(((3s,5s,7s)-adamantan-1-yl)amino)-3-oxopropyl)thio)propan-2-yl)thio)propenamide (Ad-TK-Ad). Poly-CD was assembled with CD to improve the stability of the reactor. The MNZ nanotheranostic platform can release Cu(II) and Mn(II), which could react with intracellular GSH to consume the reducing substances in tumors. Subsequently, H2O2 can be converted into •OH, and the effect is improved with increasing temperatures. Cytotoxicity of MNZ (200 μg mL-1) was studied by cell counting kit-8 (CCK-8) assay using HeLa cells as the models. Results indicated that cell viability was clearly reduced to 22% by the nanoparticles alone, to 18% by the nanoparticles with H2O2, and to 9% by the nanoparticles with H2O2 and NIR, under weak acidic condition (pH 6.8). This work provides a beneficial exploration for the application of nano-delivery strategies for combined photothermal and chemodynamic therapy agents.
Collapse
Affiliation(s)
- Fuli Lin
- School of Chemistry and Chemical Engineering, Shihezi University/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Beisi Road, Shihezi City, Xinjiang, 832003, P. R. China.
| | - Yuchang Qin
- School of Chemistry and Chemical Engineering, Shihezi University/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Beisi Road, Shihezi City, Xinjiang, 832003, P. R. China.
| | - Jingjing Sun
- School of Medicine, Shihezi University, P. R. China.
| | - Yijun Liu
- School of Chemistry and Chemical Engineering, Shihezi University/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Beisi Road, Shihezi City, Xinjiang, 832003, P. R. China.
| | - Shengchao Yang
- School of Chemistry and Chemical Engineering, Shihezi University/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Beisi Road, Shihezi City, Xinjiang, 832003, P. R. China.
| | - Shuang Zheng
- School of Chemistry and Chemical Engineering, Shihezi University/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Beisi Road, Shihezi City, Xinjiang, 832003, P. R. China.
| | - Lisha Yin
- School of Medicine, Shihezi University, P. R. China.
| | - Dongmei Li
- School of Medicine, Shihezi University, P. R. China.
| | - Lin Cui
- School of Medicine, Shihezi University, P. R. China.
| | - Gang Li
- The First Affiliated Hospital of Shihezi University, P. R. China
| | - Zhongpeng Qiu
- The First Affiliated Hospital of Shihezi University, P. R. China
| | - Zhiyong Liu
- School of Chemistry and Chemical Engineering, Shihezi University/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Beisi Road, Shihezi City, Xinjiang, 832003, P. R. China.
| |
Collapse
|
36
|
Li X, Wang H, Li Z, Liu S, Chen Y, Ruan Z, Yao Z, Wei G, Cao C, Zheng W, Guan W. Full-active pharmaceutical ingredient nanosensitizer for augmented photoimmunotherapy by synergistic mitochondria targeting and immunogenic death inducing. MedComm (Beijing) 2024; 5:e756. [PMID: 39525955 PMCID: PMC11550090 DOI: 10.1002/mco2.756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/29/2024] [Accepted: 09/01/2024] [Indexed: 11/16/2024] Open
Abstract
The precise and effective activation of the immune response is crucial in promising therapy curing cancer. Photoimmunotherapy (PIT) is an emerging strategy for precise regulation and highly spatiotemporal selectivity. However, this approach faces a significant challenge due to the off-target effect and the immunosuppressive microenvironment. To address this challenge, a nanoscale full-active pharmaceutical ingredient (API) photo-immune stimulator was developed. This formulation overcomes the limitations of PIT by strengthening the ability to penetrate tumors deeply and inducing precise and potent mitochondria-targeted dual-mode photodynamic therapy and photothermal therapy. Along with inhibiting overexpressed Hsp90, this nanosensitizer in turn improves the immunosuppressive microenvironment. Ultimately, this mitochondria-targeted PIT demonstrated potent antitumor efficacy, achieving a remarkable inhibition rate of ≥95% for both established primary tumors and distant abscopal tumors. In conclusion, this novel self-delivery full-API nanosystem enhances the efficacy of phototherapy and reprograms the immunosuppressive microenvironment, thereby holding great promise in the development of precise and effective immunotherapy.
Collapse
Affiliation(s)
- Xianghui Li
- Department of Dermatology and VenereologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
- Department of Gastrointestinal SurgeryAffiliated Nanjing Drum Tower HospitalNanjing University Medical SchoolNanjingChina
| | - Haoran Wang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent ManufactureNanjing University of Chinese MedicineNanjingChina
| | - Zhiyan Li
- Department of Gastrointestinal SurgeryAffiliated Nanjing Drum Tower HospitalNanjing University Medical SchoolNanjingChina
| | - Song Liu
- Department of Gastrointestinal SurgeryAffiliated Nanjing Drum Tower HospitalNanjing University Medical SchoolNanjingChina
| | - Yuanyuan Chen
- Department of Dermatology and VenereologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Zhuren Ruan
- Department of Dermatology and VenereologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Zhijian Yao
- Department of Dermatology and VenereologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Gao Wei
- Department of Dermatology and VenereologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Cunwei Cao
- Department of Dermatology and VenereologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Wenjun Zheng
- Department of Dermatology and VenereologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Wenxian Guan
- Department of Gastrointestinal SurgeryAffiliated Nanjing Drum Tower HospitalNanjing University Medical SchoolNanjingChina
| |
Collapse
|
37
|
Wang X, Yang Y, Wang P, Li Q, Gao W, Sun Y, Tian G, Zhang G, Xiao J. Oxygen self-supplying nanoradiosensitizer activates cGAS-STING pathway to enhance radioimmunotherapy of triple negative breast cancer. J Control Release 2024; 376:S0168-3659(24)00722-3. [PMID: 39490535 DOI: 10.1016/j.jconrel.2024.10.049] [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: 07/09/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Radiotherapy (RT)-mediated immune activation is insufficient for effective therapy of triple-negative breast cancer (TNBC) due to the immunosuppressive tumor microenvironment. Herein, we developed an oxygen self-supplying nanoradiosensitizer to activate immunogenic cell death (ICD) and the cGAS-STING signaling pathway, elevating the anti-tumor immune response and improving radioimmunotherapy for TNBC. The nanoradiosensitizer was fabricated using astragaloside liposome-encapsulated FePt alloy and MnO nanocrystals (ALFM). The ALFM targeted the glucose transporter-1 (GLUT-1) receptor in TNBC and effectively entered tumor cells. Subsequently, the ALFM responded to the weakly acidic tumor microenvironment and degraded, releasing FePt and Mn2+ ions. The released Mn2+ ions not only elevated cellular ROS levels via a Fenton-like reaction but also activated the cGAS-STING signaling pathway, which stimulated the anti-tumor immune response. In addition, the FePt alloy catalyzed a cascade reaction, producing ROS and O2 in tumor cells, alleviating tumor hypoxia, and enhancing the RT effect. Besides, ROS-mediated cell damage induced the ICD effect in TNBC, promoted dendritic cell maturation and the infiltration of cytotoxic T lymphocytes, ultimately eliciting cancer immunotherapy. In vivo experimental results demonstrated that ALFM effectively activated the antitumor immune response and improved the radioimmunotherapy effect for TNBC. Overall, this work presents an effective strategy for enhanced radioimmunotherapy of TNBC. Subsequently, the ALFM responded to weak acidic tumor microenvironment, and then degraded along with the release of FePt and Mn2+ ions. The released Mn2+ ions not only elevated cellular ROS level via Fenton-like reaction, but also activated cGAS-STING signal pathway, which activated anti-tumor immune response. In addition, FePt alloy catalyzed cascade reaction and then produced ROS and O2 in tumor cells, relieving tumor hypoxia and enhancing RT effect. Besides, ROS-mediated cell damage induced ICD effect of TNBC, promoted dendritic cells maturation and the infiltration of cytotoxic T lymphocytes, eventually elicited antitumor immunotherapy. In vivo experimental results demonstrated that ALFM effectively activated antitumor immune response, improved radioimmunotherapy effect of TNBC. Overall, this work provided a complete new strategy for enhanced radioimmunotherapy of TNBC.
Collapse
Affiliation(s)
- Xiaofei Wang
- School of Pharmacy, Institute of Aging Medicine, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, PR China
| | - Yang Yang
- School of Pharmacy, Institute of Aging Medicine, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, PR China
| | - Peng Wang
- School of Pharmacy, Institute of Aging Medicine, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, PR China
| | - Qingdong Li
- School of Pharmacy, Institute of Aging Medicine, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, PR China
| | - Wenjuan Gao
- School of Pharmacy, Institute of Aging Medicine, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, PR China
| | - Yu Sun
- School of Pharmacy, Institute of Aging Medicine, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, PR China.
| | - Geng Tian
- School of Pharmacy, Institute of Aging Medicine, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, PR China.
| | - Guilong Zhang
- School of Pharmacy, Institute of Aging Medicine, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, PR China.
| | - Jianmin Xiao
- School of Pharmacy, Institute of Aging Medicine, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, PR China.
| |
Collapse
|
38
|
Huang Z, Huang S, Song S, Ding Y, Zhou H, Zhang S, Weng L, Zhang Y, Hu Y, Yuan A, Dai Y, Luo Z, Wang L. Two-dimensional coordination risedronate-manganese nanobelts as adjuvant for cancer radiotherapy and immunotherapy. Nat Commun 2024; 15:8692. [PMID: 39375342 PMCID: PMC11458765 DOI: 10.1038/s41467-024-53084-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024] Open
Abstract
The irradiated tumor itself represents an opportunity to establish endogenous in situ vaccines. However, such in situ cancer vaccination (ISCV) triggered by radiation therapy (RT) alone is very weak and hardly elicits systemic anticancer immunity. In this study, we develop two-dimensional risedronate-manganese nanobelts (RMn-NBs) as an adjuvant for RT to address this issue. RMn-NBs exhibit good T2 magnetic resonance imaging performance and enhanced Fenton-like catalytic activity, which induces immunogenic cell death. RMn-NBs can inhibit the HIF-1α/VEGF axis to empower RT and synchronously activate the cGAS/STING pathway for promoting the secretion of type I interferon, thereby boosting RT-triggered ISCV and immune checkpoint blockade therapy against primary and metastatic tumors. RMn-NBs as a nano-adjuvant for RT show good biocompatibility and therapeutic efficacy, presenting a promising prospect for cancer radiotherapy and immunotherapy.
Collapse
Affiliation(s)
- Zhusheng Huang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
- Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, College of Optical Engineering & Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, China
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Shiqian Huang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Simin Song
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yankui Ding
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Hao Zhou
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Shaoyin Zhang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Lixing Weng
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
- Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, College of Optical Engineering & Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Ying Zhang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
- Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, College of Optical Engineering & Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| | - Zhimin Luo
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China.
- Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, College of Optical Engineering & Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, China.
| | - Lianhui Wang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China.
- Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, College of Optical Engineering & Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, China.
| |
Collapse
|
39
|
Guo Y, Li Y, Zhang M, Ma R, Wang Y, Weng X, Zhang J, Zhang Z, Chen X, Yang W. Polymeric nanocarrier via metabolism regulation mediates immunogenic cell death with spatiotemporal orchestration for cancer immunotherapy. Nat Commun 2024; 15:8586. [PMID: 39362879 PMCID: PMC11450208 DOI: 10.1038/s41467-024-53010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 09/22/2024] [Indexed: 10/05/2024] Open
Abstract
The limited efficacy of cancer immunotherapy occurs due to the lack of spatiotemporal orchestration of adaptive immune response stimulation and immunosuppressive tumor microenvironment modulation. Herein, we report a nanoplatform fabricated using a pH-sensitive triblock copolymer synthesized by reversible addition-fragmentation chain transfer polymerization enabling in situ tumor vaccination and tumor-associated macrophages (TAMs) polarization. The nanocarrier itself can induce melanoma immunogenic cell death (ICD) via tertiary amines and thioethers concentrating on mitochondria to regulate metabolism in triggering endoplasmic reticulum stress and upregulating gasdermin D for pyroptosis as well as some features of ferroptosis and apoptosis. After the addition of ligand cyclic arginine-glycine-aspartic acid (cRGD) and mannose, the mixed nanocarrier with immune adjuvant resiquimod encapsulation can target B16F10 cells for in situ tumor vaccination and TAMs for M1 phenotype polarization. In vivo studies indicate that the mixed targeting nanoplatform elicits tumor ICD, dendritic cell maturation, TAM polarization, and cytotoxic T lymphocyte infiltration and inhibits melanoma volume growth. In combination with immune checkpoint blockade, the survival time of mice is markedly prolonged. This study provides a strategy for utilizing immunoactive materials in the innate and adaptive immune responses to augment cancer therapy.
Collapse
Affiliation(s)
- Yichen Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yongjuan Li
- The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengzhe Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Rong Ma
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yayun Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xiao Weng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jinjie Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Xiaoyuan Chen
- 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, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, Singapore.
| | - Weijing Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan Province, China.
| |
Collapse
|
40
|
Chu X, Hou HY, Duan MD, Zhang YJ, Zhu YY, Liu Y, Li SL. Tumor Microenvironment Specific Regulation Ca-Fe-Nanospheres for Ferroptosis-Promoted Domino Synergistic Therapy and Tumor Immune Response. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312141. [PMID: 38801318 DOI: 10.1002/smll.202312141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/02/2024] [Indexed: 05/29/2024]
Abstract
Reactive oxygen species (ROS)-mediated emerging treatments exhibit unique advantages in cancer therapy in recent years. While the efficacy of ROS-involved tumor therapy is greatly restricted by complex tumor microenvironment (TME). Herein, a dual-metal CaO2@CDs-Fe (CCF) nanosphere, with TME response and regulation capabilities, are proposed to improve ROS lethal power by a multiple cascade synergistic therapeutic strategy with domino effect. In response to weak acidic TME, CCF will decompose, accompanied with intracellular Ca2+ upregulated and abundant H2O2 and O2 produced to reverse antitherapeutic TME. Then the exposed CF cores can act as both Fenton agent and sonosensitizer to generate excessive ROS in the regulated TME for enhanced synergistic CDT/SDT. In combination with calcium overloading, the augmented ROS induced oxidative stress will cause more severe mitochondrial damage and cellular apoptosis. Furthermore, CCF can also reduce GPX4 expression and enlarge the lipid peroxidation, causing ferroptosis and apoptosis in parallel. These signals of damage will finally initiate damage-associated molecular patterns to activate immune response and to realize excellent antitumor effect. This outstanding domino ROS/calcium loading synergistic effect endows CCF with excellent anticancer effect to efficiently eliminate tumor by apoptosis/ferroptosis/ICD both in vitro and in vivo.
Collapse
Affiliation(s)
- Xu Chu
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE), School of Material Science and Engineering & School of Chemistry, Tiangong University, Tianjin, 300387, P. R. China
| | - Hua-Ying Hou
- School of Electronic and Information Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin, 300387, P. R. China
| | - Meng-Die Duan
- School of Electronic and Information Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin, 300387, P. R. China
| | - Yu-Juan Zhang
- School of Electronic and Information Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin, 300387, P. R. China
| | - Yu-Ying Zhu
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE), School of Material Science and Engineering & School of Chemistry, Tiangong University, Tianjin, 300387, P. R. China
| | - Yi Liu
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE), School of Material Science and Engineering & School of Chemistry, Tiangong University, Tianjin, 300387, P. R. China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan, 430023, P. R. China
- Hubei Key Laboratory of Radiation Chemistry and Functional Materials, School of Nuclear Technology and Chemistry and Biology, Hubei University of Science and Technology, Xianning, 437100, China
| | - Shu-Lan Li
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE), School of Material Science and Engineering & School of Chemistry, Tiangong University, Tianjin, 300387, P. R. China
| |
Collapse
|
41
|
Shi M, Wang Y, Yang H, Lai C, Yu J, Sun Y. High doses of radiation cause cochlear immunological stress and sensorineural hearing loss. Heliyon 2024; 10:e37223. [PMID: 39309931 PMCID: PMC11414509 DOI: 10.1016/j.heliyon.2024.e37223] [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: 04/16/2024] [Revised: 08/02/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Radiotherapy is a crucial treatment for head and neck malignancies, but it can sometimes cause sensorineural hearing loss (SNHL). Changes in the immune microenvironment and sensory neuroepithelium of the inner ear after radiation exposure remain poorly understood. This study investigated cochlear morphology and macrophages in the inner ear after high-dose irradiation. The heads of heterozygous 8-week-old Cx3cr1GFP/+ male mice were irradiated with 30Gy of X-rays and biological samples were collected on days 1, 7, and 10 after irradiation. Auditory brainstem responses were used to assess auditory function in the mice. Changes in basilar membrane hair cells, spiral ganglion neurons (SGN), and inner ear macrophages were observed using hematoxylin-eosin (HE) staining and immunofluorescence staining. The expression of inflammatory mediators in the inner ear was detected by quantitative real-time reverse transcription-polymerase chain reaction (RT-qPCR) in cochlear tissue. The results showed no significant hair cell loss after a single high dose of radiation. However, the mice developed pantonal hearing loss on day 10 when HE staining revealed SGN atrophy and immunofluorescence showed decreased neurofilament expression. The number of macrophages in the inner ear reduced over time. RT-qPCR showed that cochlear inflammatory factors and chemokines were briefly upregulated on day 1st after irradiation and then decreased over time. In conclusion, high-dose irradiation causes acute SNHL that is not associated with hair cell loss and may be related to SGN changes. Radiation-induced SNHL is associated with a reduction in cochlear macrophages and changes in the immune microenvironment, but the relationship between the two remains to be investigated.
Collapse
Affiliation(s)
- Mengwen Shi
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ye Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Huiwen Yang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chengcai Lai
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jintao Yu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
42
|
Fang L, Meng Q, Wang J, Tu Y, Qu H, Diao Y, Li W, Wen H, Fang J, Hang L, Ma P, Jiang G. Multifunctional single-component photosensitizers as metal-free ferroptosis inducers for enhanced photodynamic immunotherapy. Acta Biomater 2024; 186:383-395. [PMID: 39069112 DOI: 10.1016/j.actbio.2024.07.034] [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: 01/19/2024] [Revised: 06/07/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Immunotherapy can enhance primary tumor efficacy, restrict distant growth, and combat lung metastasis. Unfortunately, it remains challenging to effectively activate the immune response. Here, tertiary butyl, methoxy, and triphenylamine (TPA) were utilized as electron donors to develop multifunctional photosensitizers (PSs). CNTPA-TPA, featuring TPA as the donor (D) and cyano as the acceptor (A), excelled in reactive oxygen species (ROS) generation due to its smaller singlet-triplet energy gap (ΔES-T) and larger spin-orbit coupling constant (SOC). Additionally, cyano groups reacted with glutamate (Glu) and glutathione (GSH), reducing intracellular GSH levels. This not only enhanced PDT efficacy but also triggered redox dyshomeostasis-mediated ferroptosis. The positive effects of photodynamic therapy (PDT) and ferroptosis promoted immunogenic cell death (ICD) and immune activation. By further combining anti-programmed cell death protein ligand-1 (anti-PD-L1) antibody, the powerful treatments of ferroptosis-assisted photodynamic immunotherapy significantly eradicated the primary tumors, inhibited the growth of distant tumors, and suppressed lung metastasis. In this study, a three-pronged approach was realized by single-component CNTPA-TPA, which simultaneously served as metal-free ferroptosis inducers, type-I photosensitizers, and immunologic adjuvants for near-infrared fluorescence imaging (NIR FLI)-guided multimodal phototheranostics of tumor. STATEMENT OF SIGNIFICANCE: (1) CNTPA-TPA shared the smallest singlet-triplet energy gap and the largest spin-orbit coupling constant, which boosted intersystem crossing for efficient type-I photodynamic therapy (PDT); (2) Special reactions between cyano groups with glutamate and glutathione in mild conditions restricted the biosynthesis of intracellular GSH. GSH-depletion efficiently induced glutathione peroxidase 4 inactivation and lipid peroxide, resulting in ferroptosis of tumor cells; (3) The combination treatments of ferroptosis-assisted photodynamic immunotherapy induced by single-component CNTPA-TPA with the participation of anti-PD-L1 antibody resulted in increased T-cell infiltration and profound suppression of both primary and distant tumor growth, as well as lung metastasis.
Collapse
Affiliation(s)
- Laiping Fang
- Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Basic Medicine, School of Medicine, Jinan University, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Renmin Street 5625, Changchun 130012, PR China
| | - Jizhuang Wang
- College of Chemistry and Materials Science, Jinan University, Huangpu Avenue West 601, Guangzhou 510632, PR China
| | - Yike Tu
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Hong Qu
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Yanzhao Diao
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Wuming Li
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Hua Wen
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Jin Fang
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Lifeng Hang
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China.
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Renmin Street 5625, Changchun 130012, PR China.
| | - Guihua Jiang
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China.
| |
Collapse
|
43
|
Malla R, Kumari S, Ganji SP, Srilatha M, Nellipudi HR, Nagaraju GP. Reactive oxygen species of tumor microenvironment: Harnessing for immunogenic cell death. Biochim Biophys Acta Rev Cancer 2024; 1879:189154. [PMID: 39019409 DOI: 10.1016/j.bbcan.2024.189154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
The tumor microenvironment (TME) is a dynamic and complex system that undergoes continuous changes in its network architecture, notably affecting redox homeostasis. These alterations collectively shape a diverse ecosystem actively supporting tumor progression by influencing the cellular and molecular components of the TME. Despite the remarkable clinical advancements in cancer immunotherapy, its spectrum of clinical utility is limited by the altered TME and inadequate tumor immunogenicity. Recent studies have revealed that some conventional and targeted therapy strategies can augment the efficacy of immunotherapy even in patients with less immunogenic solid tumors. These strategies provoke immunogenic cell death (ICD) through the ROS-dependent liberation of damage-associated molecular patterns (DAMPs). These DAMPs recognize and bind with Pattern Recognition Receptors (PRRs) on immune cells, activating and maturing defense cells, ultimately leading to a robust antitumor immune response. The present review underscores the pivotal role of redox homeostasis in orchestrating the transition of TME from a cold to a hot phenotype and the ROS-ICD axis in immune response induction. Additionally, it provides up-to-date insights into strategies that leverage ROS generation to induce ICD. The comprehensive analysis aims to develop ROS-based effective cancer immunotherapies for less immunogenic tumors.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Seema Kumari
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Swapna Priya Ganji
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh 517502, India
| | | | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA.
| |
Collapse
|
44
|
Ming Z, Zhang Y, Song L, Chen M, Lin L, He Y, Liu W, Zhu Y, Zhang Y, Zhang G. Rare Earth Nanoprobes for Targeted Delineation of Triple Negative Breast Cancer and Enhancement of Radioimmunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309992. [PMID: 38774946 PMCID: PMC11304243 DOI: 10.1002/advs.202309992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/20/2024] [Indexed: 08/09/2024]
Abstract
Radiotherapy demonstrates a synergistic effect with immunotherapy by inducing a transformation of "immune cold" tumors into "immune hot" tumors in triple negative breast cancer (TNBC). Nevertheless, the effectiveness of immunotherapy is constrained by low expression of tumor-exposed antigens, inadequate inflammation, and insufficient tumor infiltrating lymphocyte (TILs). To address this predicament, novel lutecium-based rare earth nanoparticles (RENPs) are synthesized with the aim of amplifying radiation effect and tumor immune response. The nanoprobe is characterized by neodymium-based down-conversion fluorescence, demonstrating robust photostability, biocompatibility, and targetability. The conjugation of RENPs with a CXCR4 targeted drug enables precise delineation of breast tumors using a near-infrared imaging system and improves radiation efficacy via lutetium-based radio-sensitizer in vivo. Furthermore, the study shows a notable enhancement of immune response through the induction of immunogenic cell death and recruitment of TILs, resulting in the inhibition of tumor progression both in vitro and in vivo models following the administration of nanoparticles. Hence, the novel multifunctional nanoprobes incorporating various lanthanide elements offer the potential for imaging-guided tumor delineation, radio-sensitization, and immune activation post-radiation, thus presenting an efficient radio-immunotherapeutic approach for TNBC.
Collapse
Affiliation(s)
- Zi‐He Ming
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen University School of MedicineXiamen UniversityXiamenFujian361104China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast CancerXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Key Laboratory of Endocrine‐Related Cancer Precision MedicineXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid CancersXiamenFujian361102China
| | - Yong‐Qu Zhang
- Department of Breast CenterCancer Hospital of Shantou University Medical CollegeShantouGuangdong515041China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast CancerXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Key Laboratory of Endocrine‐Related Cancer Precision MedicineXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
| | - Liang Song
- State Key Laboratory of Structural ChemistryFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhouFujian350000China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional MaterialsXiamen Institute of Rare Earth MaterialsHaixi InstituteChinese Academy of SciencesXiamenFujian361021China
| | - Min Chen
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast CancerXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Key Laboratory of Endocrine‐Related Cancer Precision MedicineXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
| | - Lin‐Ling Lin
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen University School of MedicineXiamen UniversityXiamenFujian361104China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast CancerXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Key Laboratory of Endocrine‐Related Cancer Precision MedicineXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid CancersXiamenFujian361102China
| | - Yue‐Yang He
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen University School of MedicineXiamen UniversityXiamenFujian361104China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast CancerXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Key Laboratory of Endocrine‐Related Cancer Precision MedicineXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid CancersXiamenFujian361102China
| | - Wan‐Ling Liu
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen University School of MedicineXiamen UniversityXiamenFujian361104China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast CancerXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Key Laboratory of Endocrine‐Related Cancer Precision MedicineXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid CancersXiamenFujian361102China
| | - Yuan‐Yuan Zhu
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen University School of MedicineXiamen UniversityXiamenFujian361104China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast CancerXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Key Laboratory of Endocrine‐Related Cancer Precision MedicineXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid CancersXiamenFujian361102China
| | - Yun Zhang
- State Key Laboratory of Structural ChemistryFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhouFujian350000China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional MaterialsXiamen Institute of Rare Earth MaterialsHaixi InstituteChinese Academy of SciencesXiamenFujian361021China
| | - Guo‐Jun Zhang
- Cancer Center and Department of Breast and Thyroid SurgeryXiang'an Hospital of Xiamen University School of MedicineXiamen UniversityXiamenFujian361104China
- The Breast CenterYunnan Cancer HospitalThe Third Affiliated Hospital of Kunming Medical UniversityBeijing University Cancer HospitalKunmingYunnan650118China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast CancerXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Key Laboratory of Endocrine‐Related Cancer Precision MedicineXiang'an Hospital of Xiamen UniversityXiamenFujian361102China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid CancersXiamenFujian361102China
| |
Collapse
|
45
|
Zhou Z, Jiang X, Yi L, Li C, Wang H, Xiong W, Li Z, Shen J. Mitochondria Energy Metabolism Depression as Novel Adjuvant to Sensitize Radiotherapy and Inhibit Radiation Induced-Pulmonary Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401394. [PMID: 38715382 PMCID: PMC11234447 DOI: 10.1002/advs.202401394] [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: 02/07/2024] [Indexed: 07/11/2024]
Abstract
Currently, the typical combination therapy of programmed death ligand-1 (PD-L1) antibodies with radiotherapy (RT) still exhibits impaired immunogenic antitumor response in clinical due to lessened DNA damage and acquired immune tolerance via the upregulation of some other immune checkpoint inhibitors. Apart from this, such combination therapy may raise the occurrence rate of radiation-induced lung fibrosis (RIPF) due to enhanced systemic inflammation, leading to the ultimate death of cancer patients (average survival time of about 3 years). Therefore, it is newly revealed that mitochondria energy metabolism regulation can be used as a novel effective PD-L1 and transforming growth factor-β (TGF-β) dual-downregulation method. Following this, IR-TAM is prepared by conjugating mitochondria-targeted heptamethine cyanine dye IR-68 with oxidative phosphorylation (OXPHOS) inhibitor Tamoxifen (TAM), which then self-assembled with albumin (Alb) to form IR-TAM@Alb nanoparticles. By doing this, tumor-targeting IR-TAM@Alb nanoparticle effectively reversed tumor hypoxia and depressed PD-L1 and TGF-β expression to sensitize RT. Meanwhile, due to the capacity of heptamethine cyanine dye in targeting RIPF and the function of TAM in depressing TGF-β, IR-TAM@Alb also ameliorated fibrosis development induced by RT.
Collapse
Affiliation(s)
- Zaigang Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xin Jiang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lei Yi
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Cheng Li
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Haoxiang Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wei Xiong
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Zhipeng Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| |
Collapse
|
46
|
Zhu D, Lu Y, Yang S, Hu T, Tan C, Liang R, Wang Y. PAD4 Inhibitor-Functionalized Layered Double Hydroxide Nanosheets for Synergistic Sonodynamic Therapy/Immunotherapy Of Tumor Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401064. [PMID: 38708711 PMCID: PMC11234469 DOI: 10.1002/advs.202401064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/03/2024] [Indexed: 05/07/2024]
Abstract
Sonodynamic therapy (SDT) is demonstrated to trigger the systemic immune response of the organism and facilitate the treatment of metastatic tumors. However, SDT-mediated neutrophil extracellular traps (NETs) formation can promote tumor cell spread, thus weakening the therapeutic effectiveness of metastatic tumors. Herein, the amorphous CoW-layered double hydroxide (a-CoW-LDH) nanosheets are functionalized with a peptidyl arginine deiminase 4 (PAD4) inhibitor, i.e., YW3-56, to construct a multifunctional nanoagent (a-LDH@356) for synergistic SDT/immunotherapy. Specifically, a-CoW-LDH nanosheets can act as a sonosensitizer to generate abundant reactive oxygen species (ROS) under US irradiation. After loading with YW3-56, a-LDH@356 plus US irradiation not only effectively induces ROS generation and immunogenic cell death, but also inhibits the elevation of citrullinated histone H3 (H3cit) and the release of NETs, enabling a synergistic enhancement of anti-tumor metastasis effect. Using 4T1 tumor model, it is demonstrated that combining a-CoW-LDH with YW3-56 stimulates an anti-tumor response by upregulating the proportion of immune-activated cells and inducing polarization of M1 macrophages, and inhibits immune escape by downregulating the expression of PD-1 on immune cells under US irradiation, which not only arrests primary tumor progression with a tumor inhibition rate of 69.5% but also prevents tumor metastasis with the least number of lung metastatic nodules.
Collapse
Affiliation(s)
- Di Zhu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing, 100069, P. R. China
| | - Yu Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing, 100069, P. R. China
| | - Shuqing Yang
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Tingting Hu
- Department Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, 999077, P. R. China
| | - Chaoliang Tan
- Department Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, 999077, P. R. China
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
- Quzhou Institute for Innovation in Resource Chemical Engineering, Quzhou, 324000, P. R. China
| | - Yuji Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing, 100069, P. R. China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Laboratory for Clinical Medicine, Capital Medical University, Beijing Laboratory of Oral Health, Beijing, 100069, P. R. China
| |
Collapse
|
47
|
Huang Z, Song J, Huang S, Wang S, Shen C, Song S, Lian J, Ding Y, Gong Y, Zhang Y, Yuan A, Hu Y, Tan C, Luo Z, Wang L. Phase and Defect Engineering of MoSe 2 Nanosheets for Enhanced NIR-II Photothermal Immunotherapy. NANO LETTERS 2024; 24:7764-7773. [PMID: 38864366 DOI: 10.1021/acs.nanolett.4c01879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
Inducing immunogenic cell death (ICD) during photothermal therapy (PTT) has the potential to effectively trigger photothermal immunotherapy (PTI). However, ICD induced by PTT alone is often limited by inefficient PTT, low immunogenicity of tumor cells, and a dysregulated redox microenvironment. Herein, we develop MoSe2 nanosheets with high-percentage metallic 1T phase and rich exposed active Mo centers through phase and defect engineering of MoSe2 as an effective nanoagent for PTI. The metallic 1T phase in MoSe2 nanosheets endows them with strong PTT performance, and the abundant exposed active Mo centers endow them with high activity for glutathione (GSH) depletion. The MoSe2-mediated high-performance PTT synergizing with efficient GSH depletion facilitates the release of tumor-associated antigens to induce robust ICD, thus significantly enhancing checkpoint blockade immunotherapy and activating systemic immune response in mouse models of colorectal cancer and triple-negative metastatic breast cancer.
Collapse
Affiliation(s)
- Zhusheng Huang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, People's Republic of China
- Faculty of Health Sciences and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, 999078, People's Republic of China
| | - Jingrun Song
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, People's Republic of China
| | - Shiqian Huang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, People's Republic of China
| | - Shengheng Wang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, People's Republic of China
| | - Chuang Shen
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, People's Republic of China
| | - Simin Song
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, People's Republic of China
| | - Jianhui Lian
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, People's Republic of China
| | - Yankui Ding
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, People's Republic of China
| | - Yue Gong
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
| | - Ying Zhang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, People's Republic of China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, People's Republic of China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, People's Republic of China
| | - Chaoliang Tan
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong SAR 999077, People's Republic of China
| | - Zhimin Luo
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, People's Republic of China
| | - Lianhui Wang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, People's Republic of China
| |
Collapse
|
48
|
Peng J, Li S, Ti H. Sensitize Tumor Immunotherapy: Immunogenic Cell Death Inducing Nanosystems. Int J Nanomedicine 2024; 19:5895-5930. [PMID: 38895146 PMCID: PMC11184231 DOI: 10.2147/ijn.s457782] [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: 01/28/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Low immunogenicity of tumors poses a challenge in the development of effective tumor immunotherapy. However, emerging evidence suggests that certain therapeutic approaches, such as chemotherapy, radiotherapy, and phototherapy, can induce varying degrees of immunogenic cell death (ICD). This ICD phenomenon leads to the release of tumor antigens and the maturation of dendritic cells (DCs), thereby enhancing tumor immunogenicity and promoting immune responses. However, the use of a single conventional ICD inducer often fails to achieve in situ tumor ablation and establish long-term anti-tumor immune responses. Furthermore, the induction of ICD induction varies among different approaches, and the distribution of the therapeutic agent within the body influences the level of ICD and the occurrence of toxic side effects. To address these challenges and further boost tumor immunity, researchers have explored nanosystems as inducers of ICD in combination with tumor immunotherapy. This review examines the mechanisms of ICD and different induction methods, with a specific focus on the relationship between ICD and tumor immunity. The aim is to explore the research advancements utilizing various nanomaterials to enhance the body's anti-tumor effects by inducing ICD. This paper aims to contribute to the development and clinical application of nanomaterial-based ICD inducers in the field of cancer immunotherapy by providing important theoretical guidance and practical references.
Collapse
Affiliation(s)
- Jianlan Peng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Shiying Li
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Huihui Ti
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- Guangdong Province Precise Medicine and Big Data Engineering Technology Research Center for Traditional Chinese Medicine, Guangzhou, People’s Republic of China
| |
Collapse
|
49
|
Li R, Zhao W, Han Z, Feng N, Wu T, Xiong H, Jiang W. Self-Cascade Nanozyme Reactor as a Cuproptosis Inducer Synergistic Inhibition of Cellular Respiration Boosting Radioimmunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306263. [PMID: 38221757 DOI: 10.1002/smll.202306263] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/08/2023] [Indexed: 01/16/2024]
Abstract
Intrinsic or acquired radioresistance remained an important challenge in the successful management of cancer. Herein, a novel "smart" multifunctional copper-based nanocomposite (RCL@Pd@CuZ) to improve radiotherapy (RT) sensitivity is designed and developed. In this nanoplatform, DSPE-PEG-RGD modified on the liposome surface enhanced tumor targeting and permeability; capsaicin inserted into the phospholipid bilayer improved the hypoxic conditions in the tumor microenvironment (TME) by inhibiting mitochondrial respiration; a Cu MOF porous cube encapsulated in liposome generated highly active hydroxyl radicals (OH·), consumed GSH and promoted cuproptosis by releasing Cu2+; the ultrasmall palladium (Pd) nanozyme within the cubes exhibited peroxidase activity, catalyzing toxic OH· generation and releasing oxygen from hydrogen peroxide; and lastly, Pd, as an element with a relatively high atomic number (Z) enhanced the photoelectric and Compton effects of X-rays. Therefore, RCL@Pd@CuZ enhance RT sensitivity by ameliorating hypoxia, promoting cuproptosis, depleting GSH, amplifying oxidative stress, and enhancing X-ray absorption , consequently potently magnifying immunogenic cell death (ICD). In a mouse model , RCL@Pd@CuZ combined with RT yielded >90% inhibition compared with that obtained by RT alone in addition to a greater quantity of DC maturation and CD8+ T cell infiltration. This nanoplatform offered a promising remedial modality to facilitate cuproptosis-related cancer radioimmunotherapy.
Collapse
Affiliation(s)
- Rui Li
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430000, China
- Department of Respiratory Intervention, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Jinshui, Zhengzhou, 450008, China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Zhuo Han
- Department of General Surgery, Tangdu Hospital, the Air Force Medical University, Xi'an, 710000, China
| | - Na Feng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Tingting Wu
- Nanozyme Medical Center, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, China
- Department of Pharmacy of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Wei Jiang
- Nanozyme Medical Center, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, China
- Department of Pharmacy of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
50
|
Qiu J, Cheng Z, Jiang Z, Gan L, Zhang Z, Xie Z. Immunomodulatory Precision: A Narrative Review Exploring the Critical Role of Immune Checkpoint Inhibitors in Cancer Treatment. Int J Mol Sci 2024; 25:5490. [PMID: 38791528 PMCID: PMC11122264 DOI: 10.3390/ijms25105490] [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: 03/24/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
An immune checkpoint is a signaling pathway that regulates the recognition of antigens by T-cell receptors (TCRs) during an immune response. These checkpoints play a pivotal role in suppressing excessive immune responses and maintaining immune homeostasis against viral or microbial infections. There are several FDA-approved immune checkpoint inhibitors (ICIs), including ipilimumab, pembrolizumab, and avelumab. These ICIs target cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed death ligand 1 (PD-L1). Furthermore, ongoing efforts are focused on developing new ICIs with emerging potential. In comparison to conventional treatments, ICIs offer the advantages of reduced side effects and durable responses. There is growing interest in the potential of combining different ICIs with chemotherapy, radiation therapy, or targeted therapies. This article comprehensively reviews the classification, mechanism of action, application, and combination strategies of ICIs in various cancers and discusses their current limitations. Our objective is to contribute to the future development of more effective anticancer drugs targeting immune checkpoints.
Collapse
Affiliation(s)
- Junyu Qiu
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zilin Cheng
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zheng Jiang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Luhan Gan
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Huan Kui School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zixuan Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
| |
Collapse
|