1
|
Lin Q, Zhang Y, Zeng Y, Zha Y, Xue W, Yu S. Hybrid membrane based biomimetic nanodrug with high-efficient melanoma-homing and NIR-II laser-amplified peroxynitrite boost properties for enhancing antitumor therapy via effective immunoactivation. Biomaterials 2025; 317:123045. [PMID: 39742839 DOI: 10.1016/j.biomaterials.2024.123045] [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: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/04/2025]
Abstract
Owing to the excellent stability, anticancer activity and immunogenicity, peroxynitrite (ONOO-) has been gained enormous interests in cancer therapy. Nevertheless, precise delivery and control release of ONOO- in tumors remains a big challenge. Herein, B16F10 cancer cell membrane/liposome hybrid membrane (CM-Lip) based biomimetic nanodrug with high-efficient tumor-homing and NIR-II laser controlled ONOO- boost properties was designed for melanoma treatment. Briefly, NIR-II molecule IR1061, NO donor BNN6 and β-lapachone (Lapa) were firstly encapsulated in the heat-responsive palmitoyl phosphatidylcholine/cholesterol liposome, followed by fusion with B16F10 cell membrane (CM) to obtain biomimetic CM-Lip@(IR/BNN6/Lapa). The hybrid membrane-based nanodrug displayed excellent biocompatibility and melanoma-targeting efficiency. Upon 1064 nm laser irradiation, the mild photothermal effect of CM-Lip@(IR/BNN6/Lapa) firstly triggered the release of NO and Lapa, which subsequently catalyzed the quinone oxidoreductase 1 (NQO1) overexpressed in tumors to produce O2•-, finally caused intraturmal ONOO- boost via cascade reaction. The boosted ONOO- could effectively inhibit melanoma by ways of triggering mitochondrion-mediated apoptotic pathway, upregulating 3-nitrotyrosine expression, inducing DNA damage and inhibiting DNA repair enzyme expression of poly (ADP-ribose) polymerase 1 (PARP-1). Moreover, ONOO- displayed excellent immunoactivation and immunomodulation activities by effectively inducing immunogenic tumor cell death, promoting dendritic cells maturation, increasing cytotoxic T lymphocytes expression and repolarizing M1-phenotype macrophages.
Collapse
Affiliation(s)
- Qi Lin
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Yu Zhang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Yina Zeng
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Yongchao Zha
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Siming Yu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
2
|
Yang C, Liao X, Zhou K, Yao Y, He X, Zhong W, Zheng D, Yang Y, Li M, Zhou M, Zhou Y, Li L, Bai Y, Shi K, Qian Z. Multifunctional nanoparticles and collagenase dual loaded thermosensitive hydrogel system for enhanced tumor-penetration, reversed immune suppression and photodynamic-immunotherapy. Bioact Mater 2025; 48:1-17. [PMID: 40028237 PMCID: PMC11870144 DOI: 10.1016/j.bioactmat.2025.02.014] [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: 12/03/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 03/05/2025] Open
Abstract
Breast cancer is the most prevalent and lethal malignancy among females, with a critical need for safer and less invasive treatments. Photodynamic therapy (PDT) can effectively eliminate tumor cells with minimal side effects. Furthermore, the combination of PDT and immunotherapy using nanoparticles has shown promise in treating both primary and distant metastatic tumor cells. Therefore, this study proposes applying the PDT-immunotherapy combination to breast cancer treatment. However, the low immunogenicity characteristic of "cold" tumors in part of breast cancer significantly diminishes therapeutic efficacy. To address this challenge, here, a nano-gel system (designated as HCSC-gel) is constructed, which co-delivers a mitochondria-targeted photosensitizer and a STING agonist, capable of robustly activating "cold" tumor immunity. This system is further enhanced by collagenase (CN) to improve therapeutic outcomes. Upon injection into the primary tumor site, HCSC-gel rapidly forms a gel matrix, releasing CN to degrade the tumor extracellular matrix and facilitate the penetration of photosensitizers, STING agonists, and oxygen into the tumor tissue. Under laser irradiation, PDT and STING-mediated immune responses are activated, reversing the low immunogenicity of breast cancer and effectively treating both primary and metastatic lesions. This HCSC-gel nano hydrogel delivery platform is anticipated to provide novel insights for the clinical management of breast cancer and other low immunogenic "cold" tumors, offering significant benefits to patients.
Collapse
Affiliation(s)
- Chengli Yang
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Xukun Liao
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Kai Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Yongchao Yao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Xinlong He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Wen Zhong
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Dan Zheng
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yan Yang
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Ming Li
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Meng Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yadi Zhou
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Lin Li
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yang Bai
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| |
Collapse
|
3
|
Weissenstein U, Tschumi S, Leonhard B, Baumgartner S. A fermented Mistletoe (Viscum album L.) extract elicits markers characteristic for immunogenic cell death driven by endoplasmic reticulum stress in vitro. BMC Complement Med Ther 2025; 25:175. [PMID: 40369535 PMCID: PMC12076857 DOI: 10.1186/s12906-025-04909-8] [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: 11/12/2024] [Accepted: 05/01/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Immune evasion is a characteristic hallmark of cancer. Immunotherapies aim to activate and support the body's immune system to recognize and fight tumor cells. Induction of immunogenic cell death (ICD) and the associated activation of danger signaling pathways can increase the immunogenicity of tumor cells. Therapeutic ICD stimuli activate endoplasmic reticulum stress pathways and apoptosis leading to the cellular expression of damage-associated molecular patterns (DAMPs). The aim of our in vitro study was to investigate whether mistletoe extracts induce characteristics of immunogenic tumor cell death in cancer cell lines. METHODS Three human breast cancer cell lines and one murine melanoma cell line (SKBR3, MDA-MB-231, MCF-7, and B16F10) were treated with aqueous, fermented Viscum album extract (VAE: Iscador Qu spec.) and taxol or tunicamycin as positive controls, respectively. To investigate whether VAE induces ribotoxic stress, we measured the ER stress regulators p-eIF2a, ATF4, and CHOP by Western blot. Cell surface exposure of DAMPs (calreticulin, heat shock proteins hsp70 and hsp90), apoptosis and induction of mitochondrial reactive oxygen species (ROS) were assessed by flow cytometry. HMGB1 and ATP were quantified by ELISA and chemiluminescence assay, respectively. RESULTS Treatment with VAE resulted in phosphorylation of eIF2α in all cancer cell lines tested and increased calreticulin (CRT) exposure on the surface of pre-apoptotic SKBR3 breast cancer and B16F10 mouse melanoma cells. VAE exerted a concentration-dependent effect in all cell lines, resulting in a significantly increased exposure of three DAMPs (CRT, hsp70 and hsp90) on the surface of early apoptotic cells. Furthermore, VAE elevated mitochondrial ROS production and the release of ATP. HMGB1 release was not induced by VAE. CONCLUSIONS In this in vitro study, we demonstrated for the first time the potential of a mistletoe extract to induce surrogate markers of immunogenic cancer cell death. This is a primary step in investigating the potential of VAEs to contribute to ICD-induced tumor-specific immune activation.
Collapse
Affiliation(s)
| | | | | | - Stephan Baumgartner
- Society for Cancer Research, Arlesheim, Switzerland
- Institute of Integrative Medicine, Witten/Herdecke University, Herdecke, Germany
| |
Collapse
|
4
|
Omer S, Mambili-Mamboundou H. Analysis of a combination of cancer treatments in efforts to overcome drug resistance. Comput Biol Med 2025; 192:110278. [PMID: 40359680 DOI: 10.1016/j.compbiomed.2025.110278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
Tumor heterogeneity, the variability among cancer cells within a tumor, is a major contributor to drug resistance and poses challenges to effective treatment. To address this, we developed a mathematical model that captures tumor-immune interactions and the combined effects of chemotherapy and immunotherapy, focusing on their potential synergy. The model includes two tumor cell populations: chemosensitive (responsive to chemotherapy) and chemoresistant (unresponsive). We analyzed the stability of equilibrium states under three treatment scenarios: no treatment, chemotherapy alone, and combined therapy. To identify key parameters influencing the effectiveness of the combined treatment in reducing the overall tumor population, we performed a sensitivity analysis using Latin Hypercube Sampling (LHS) to calculate Partial Rank Correlation Coefficients (PRCCs) and their associated p-values. The analysis revealed that the killing rate of chemosensitive tumor cells by chemotherapy, the killing rate of chemosensitive tumor cells by immune cells, and the external doses of tumor-infiltrating lymphocyte (TIL) therapy were critical parameters. We formulated a quadratic optimal control problem to minimize tumor burden and treatment administration. Numerical simulations were conducted under two scenarios: with and without optimal control. Simulations without optimal control revealed that chemotherapy alone failed to eradicate tumors, while high-dose immunotherapy was more effective. However, combining the two therapies resulted in greater tumor reduction than either therapy alone. Under optimal control, our findings suggest that the most effective strategy involves administering a full chemotherapy dose along with gradually decreasing doses of TILs and interleukin-2 (IL-2).
Collapse
Affiliation(s)
- Salaheldin Omer
- University of KwaZulu Natal, School of Mathematics, Statistics, and Computer Science, Private Bag X01, Scottsville, Pietermaritzburg 3209, South Africa.
| | - Hermane Mambili-Mamboundou
- University of KwaZulu Natal, School of Mathematics, Statistics, and Computer Science, Private Bag X01, Scottsville, Pietermaritzburg 3209, South Africa
| |
Collapse
|
5
|
Tao H, Liu H, Zhang H, Ren H, Wen B, Zhang J, Du J, Cai Z, Deng Z. HGF/c-Met Axis-Targeted Nanotherapy via GSH-Responsive Polymer Platforms Suppresses Uveal Melanoma Metastasis. Adv Healthc Mater 2025:e2405056. [PMID: 40341826 DOI: 10.1002/adhm.202405056] [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: 12/14/2024] [Revised: 04/11/2025] [Indexed: 05/11/2025]
Abstract
Uveal melanoma (UM), a malignant tumor originating within the ocular, characterizes high metastasis and lethality among patients. Cancer stem cells (CSCs) distinguished by the c-Met protein are believed to mediate tumor metastasis in UM. However, the low bioavailability of c-Met inhibitors like Crizotilib (Criz) limits their clinical application. Herein, a GSH-responsive nanoparticle named NP@Oxa/Criz to precisely deliver Criz and Oxaliplatin (Oxa) is synthesized in this study. The dual-action mechanism of NP@Oxa/Criz inhibits the HGF/c-Met axis to prevent the nuclear translocation of β-Catenin, thereby reducing the transcription of metastasis-associated genes and undermining the stemness and metastasis of UM cells. Simultaneously, NP@Oxa/Criz induces immunogenic cell death to boost anti-tumor immunity. In vivo studies demonstrate that NP@Oxa/Criz can accumulate in tumor sites, significantly eradicating the primary UM in the ocular and suppressing the metastasis UM in the liver and peritoneal. The outcomes from this work illuminate the therapeutic mechanisms of NP@Oxa/Criz and provide a precise and potent nanotherapeutic strategy for clinical treatment and research in highly metastatic UM.
Collapse
Affiliation(s)
- Hui Tao
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Hanhan Liu
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Hanchen Zhang
- Chinese Academy of Sciences, Beijing, 100049, China
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Polymer Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Hong Ren
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Binyu Wen
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Jing Zhang
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Jiaqi Du
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Ziyi Cai
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhihong Deng
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| |
Collapse
|
6
|
Li Z, Ren G, Wang X, Li X, Ding L, Zhu J, Zhang Y, Zhang C, Zou J, Chen X. Tumor microenvironment responsive nano-PROTAC for BRD4 degradation enhanced cancer photo-immunotherapy. Biomaterials 2025; 322:123387. [PMID: 40344878 DOI: 10.1016/j.biomaterials.2025.123387] [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: 12/03/2024] [Revised: 04/12/2025] [Accepted: 05/01/2025] [Indexed: 05/11/2025]
Abstract
Proteolysis Targeting Chimeras (PROTAC) technology has garnered great attention due to its advantages in targeted protein degradation, promising its potential for treating malignant cancer. Nevertheless, the inherent drawbacks of PROTAC technology hinder its clinical translation. The integration of nanotechnology with PROTAC molecules to create nano-PROTACs for combined therapy offers a promising solution. Among the various cancer treatment methods, phototherapy is considered the optimal choice to integrate with specific PROTACs due to its proven effectiveness and non-invasive nature. Herein, a nano-PROTAC formulation (ARV@PEG-ICG) consisting of a phototherapeutic agent named indocyanine green functionalized polyethylene glycol (PEG-ICG) and a BRD4 degrader (ARV-825) was fabricated for cancer photo-immunotherapy. Activated by acidic tumor microenvironment (TME), ARV@PEG-ICG nanoparticles (NPs) will decompose rapidly for ARV delivery. PEG-ICG generated abundant ROS with laser irradiation, downregulating the expression of Bcl-xL and inducing the cleavage of PARP to stimulate cell apoptosis. Furthermore, the degradation of BRD4, a transcriptional cofactor, inhibited nitric oxide synthase (iNOS) generation to improve phototherapeutic efficacy. In a 4T1 breast tumor model, dying 4T1 cells released tumor associated antigens (TAAs) to serve as the immunogenic cell death (ICD) inducer, facilitating DC maturation and T cell activation and amplifying systemic immune response. The distant tumor growth can also be inhibited due to the activation of long-term immune response. Overall, the current study aims to combine typical PROTAC with functional nanomaterials to form nano-PROTAC with high performance for PROTAC delivery mediated cancer treatment.
Collapse
Affiliation(s)
- Zheng Li
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore; Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Guodong Ren
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Xuewei Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Xiaowan Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Lingwen Ding
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Jianwei Zhu
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore; Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore; Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore
| | - Yajie Zhang
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore; Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Chengwu Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China.
| | - Jianhua Zou
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore; Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore; Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore.
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore; Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore; Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore; Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Lower Kent Ridge Road, 4 Science Drive 2, 117544, Singapore.
| |
Collapse
|
7
|
Chen Y, Song Y, Zhang C, Jin P, Fu Y, Wang G, Tang L, Chen J, Xu X, Huang P. Ultrasound-responsive release of CD39 inhibitor overcomes adenosine-mediated immunosuppression in triple-negative breast cancer. J Control Release 2025; 383:113819. [PMID: 40345625 DOI: 10.1016/j.jconrel.2025.113819] [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: 03/03/2025] [Revised: 04/28/2025] [Accepted: 05/04/2025] [Indexed: 05/11/2025]
Abstract
Triple-negative breast cancer (TNBC), an exceptionally aggressive subtype of breast cancer, is characterized by a poor prognosis and limited treatment options. Although immunotherapy has shown promise for the treatment of TNBC, the immunosuppressive accumulation of adenosine (ADO) in the tumor microenvironment (TME) contributes to immune evasion and tumor progression. To address this challenge, we introduce a novel ultrasound-responsive liposomal system (BFPL) designed to inhibit ADO production and enhance the effectiveness of sonoimmunotherapy. BFPL consists of lipid membranes loaded with an endoplasmic reticulum (ER)-targeting sonosensitizer (PMPS) and a reactive oxygen species (ROS)-responsive CD39 inhibitor (FPL-67156) polyplex, synthesized via the thin-film hydration method. Upon ultrasound irradiation, BFPL generates substantial ROS, inducing robust immunogenic cell death (ICD) through ER stress. Concurrently, ROS-mediated deboronation of the polyplex releases FPL-67156, which inhibits ATP degradation into ADO, thereby promoting dendritic cell maturation and activating effector T cells. Moreover, BFPL effectively triggers a potent antitumor immune response and enhances the efficacy of anti-PD-L1 immunotherapy. Thus, by modulating metabolic pathways to counteract ADO-associated barriers in ICD therapy, this innovative approach holds potential for improving immunotherapy outcomes in TNBC.
Collapse
Affiliation(s)
- Yijie Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Ultrasound, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Yue Song
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Chao Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Peile Jin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yuhan Fu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Lina Tang
- Department of Ultrasound, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiaodan Xu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
8
|
Tian S, Xu H, Wu X, Ding Y, Liang L, Yin H, Zeng X, Liu Y, Zhu W. Ruthenium(II) polypyridyl complexes inhibit tumor growth through stimulating immune system to increase CD8 + T cell. Eur J Med Chem 2025; 289:117470. [PMID: 40054298 DOI: 10.1016/j.ejmech.2025.117470] [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: 12/23/2024] [Revised: 02/16/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025]
Abstract
In this work, we have carefully designed and synthesized two Ru(II) metal complexes: [Ru(phen)2(HMPIP)](PF6)2 (6a, where phen = 1,10-phenanthroline, HMPIP = 2-(2-hydroxy-3-methylphenyl-1H-imidazo[4,5-f][1,10]phenanthroline) and [Ru(bpy)2(HMPIP)](PF6)2 (6b, where bpy = 2,2'-bipyridine). Using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) to explore the cytotoxicity of 6a and 6b towards HepG2, B16, A549, SGC-7901, HCT116 and non-cancer LO2. The complexes exhibited cytotoxicity activity against HepG2 cells. The capacity of 6a and 6b to impede the proliferation and dissemination of cancer cells was evaluated by conducting proliferation and migration experiments and 3D model. The anticancer mechanism was investigated in detail. The utilization of cycle blocking assays revealed that 6a and 6b induced a G0/G1 phase arrest in HepG2 cells. The cellular uptake experiments show that the complexes enter the cell nuclei, then escape from the cell nuclei into the cytoplasm, finally accumulate in the mitochondria. Apoptosis assays and the examination of proteins indicated that the complexes were capable of efficiently inducing apoptosis in HepG2 cells. Additionally, the potential induction of autophagy-mediated cell death was explored. The observed reduction in glutathione (GSH) levels and glutathione peroxidase 4 (GPX4) expression suggested a disruption of redox homeostasis within cancer cells, an increment in malondialdehyde (MDA) amount, together with BODIPY staining experiment, confirm that 6a and 6b can induce ferroptosis. Interestingly, in a nude mouse model, 6a showed a significant suppression of tumor growth with an inhibition rate of 63.4 %, without causing any weight loss of mice. The studies on the mechanism show that 6a causes immune cell death, increase the amount of TNF-α and IFN-γ, reduce IL-10 content, which further activates immune response to increase CD8+ T cells to prevent tumor growth. Therefore, 6a inhibits the tumor growth through stimulating the immune response to increase CD8+ T cells. In addition, the experiments in vitro show that the complexes through inhibition of PI3K/AKT/mTOR signaling pathway and intrinsic mitochondria pathway to cause cell apoptosis. These results demonstrate that Ru(II) complexes may be potent anticancer candidates for HepG2 tumor.
Collapse
Affiliation(s)
- Shuang Tian
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Haixin Xu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xiaoyu Wu
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yueyao Ding
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Lijuan Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Hui Yin
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Xiandong Zeng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Wenrun Zhu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
9
|
Li T, Meng H, Huang X, Yu Q, Sheng S, Jiang Y, Ren F. Photodynamic Biomimetic Liposomes Targeted to the Endoplasmic Reticulum Enhance Combined Immunotherapy for Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:25112-25127. [PMID: 40258178 DOI: 10.1021/acsami.5c03687] [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: 04/23/2025]
Abstract
Cancer immunotherapies, such as immune checkpoint inhibitors, have advanced rapidly and achieved notable success, yet they face significant challenges due to poor response rates and immune-related adverse effects, particularly in cases of triple-negative breast cancer (TNBC). Photodynamic therapy (PDT) can initiate immunogenic cell death (ICD) by inducing endoplasmic reticulum (ER) stress, thereby enhancing the effectiveness of tumor immunotherapy. Herein, we develop potent PDT biomimetic liposomes (PB Lipo) locating the ER to realize a synergistic immuno-photodynamic treatment. The PB Lipo is prepared using the optimal ratios of the phospholipids in the ER membrane. It is then loaded with indocyanine green (ICG), a photosensitizer approved for clinical use. PB Lipo has the unique ability to accumulate in the ER via membrane fusion, leading to severe ER stress when exposed to near-infrared (NIR) laser light, thus intensifying ICD. In combination with the antiprogrammed death-ligand 1 (PD-L1) antibody (αPD-L1), PB Lipo significantly improves efficiency against tumors in xenograft TNBC models. As a result, our combined treatment enhances mature dendritic cells, activates CD4+ T and CD8+ T cells, and promotes the secretion of cytotoxic cytokines. Collectively, our findings reveal that PB Lipo-mediated PDT presents a viable approach for effectively targeting the ER and enhancing ICD, thereby boosting antitumor efficacy in TNBC.
Collapse
Affiliation(s)
- Tianyang Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Haimei Meng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xinfeng Huang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qin Yu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Sizhe Sheng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yufei Jiang
- First clinical medicine college, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Fei Ren
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
10
|
Huang P, Zhu J, Guo L, Zhang ZY, Guan L, Yang S. Effects of anlotinib hydrochloride on the expression of immunogenic cell death-related molecules in Cal27 tongue cancer cells. Discov Oncol 2025; 16:639. [PMID: 40299226 PMCID: PMC12040774 DOI: 10.1007/s12672-025-02464-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
OBJECTIVE To investigate the effects of anlotinib hydrochloride (AL3818) on the expression of Immunogenic Cell Death (ICD)-related molecules in tongue cancer cells. METHODS (1) The human Tongue Squamous Cell Carcinoma (TSCC) cell line Cal27 was cultured, and the half-maximal inhibitory concentration (IC50) of AL3818 and paclitaxel (PTX) on Cal27 cells was determined using the CCK-8 assay. The cells were divided into four groups: control group, AL3818 group, PTX group, and AL3818 + PTX group; (2) The apoptosis rate of each group was measured by flow cytometry; (3) The levels of calreticulin (CRT) and heat shock protein 70 (HSP70) were detected by immunofluorescence and flow cytometry; (4) The concentration of High Mobility Group Box 1 (HMGB1) was measured by Enzyme-Linked Immunosorbent Assay (ELISA); (5) Adenosine Triphosphate (ATP) levels were assessed using an ATP luminescence assay kit; and (6) The cells were divided into the control group, AL3818 group, AL3818 + CCT020312 (perk activator) treatment group, and AL3818 + ISRIB (perk inhibitor) treatment group, and detect the relative expression levels of total CRT, membrane-bound CRT, p-perk, and p-eIF2a in each group using the western blot method. RESULTS (1) The IC50 values of AL3818 and PTX at 24 h were 6.254 μmol/L and 1.718 μmol/L, respectively (P < 0.01); (2) AL3818 and PTX induced apoptosis in Cal27 cells (P < 0.05), with the highest apoptosis rate observed in the AL3818 + PTX group compared to the other three groups (P < 0.05); (3) AL3818 and PTX increased the expression of ICD-related molecules, including CRT, HMGB1, ATP, and HSP70, with the AL3818 + PTX group demonstrating the most significant effect (P < 0.05); and (4) AL3818 can induce early exposure of CRT on the membrane in the Cal27 cell line. Compared with the control group, the AL3818 + CCT020312 positive control group, and the AL3818 + ISRIB negative control group, the differences are statistically significant, while the total CRT remains roughly unchanged. The experimental results also indicate that after AL3818 acts on the Cal27 cell line for 24 h, there is an upregulation of p-perk and p-eIF2a expression along with the synchronous expression of CRT on the membrane. CONCLUSION AL3818 has the potential to induce ICD in the Cal27 TSCC cell line by modulating the levels of ICD-related molecules such as ATP, CRT, HSP70, and HMGB1. Moreover, the combination of AL3818 and PTX is more effective in inducing ICD in Cal27 cells compared to either agent alone. The process by which CRT is transferred from within the cell to the membrane during the induction of ICD in the Cal27 cell line by AL3818 may be related to the phosphorylation and activation of the PERK/elF2a signaling pathway.
Collapse
Affiliation(s)
- Pengcheng Huang
- School of Stomatology, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining, 629000, China
| | - Jitao Zhu
- Zunyi Medical University, Zunyi, 563006, Guizhou, China
| | - Lijuan Guo
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining, 629000, China
| | - Zong Yao Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, No.203 Huai Bin Road, Tian Jia'an District, Huainan, 232007, China
| | - Li Guan
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining, 629000, China
| | - Sen Yang
- School of Stomatology, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining, 629000, China.
| |
Collapse
|
11
|
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
|
12
|
Zou JX, Chang MR, Kuznetsov NA, Kee JX, Babak MV, Ang WH. Metal-based immunogenic cell death inducers for cancer immunotherapy. Chem Sci 2025; 16:6160-6187. [PMID: 40160356 PMCID: PMC11949249 DOI: 10.1039/d4sc08495k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Immunogenic cell death (ICD) has attracted enormous attention over the past decade due to its unique characteristics in cancer cell death and its role in activating innate and adaptive immune responses against tumours. Many efforts have been dedicated to screening, identifying and discovering ICD inducers, resulting in the validation of several based on metal complexes. In this review, we provide a comprehensive summary of current metal-based ICD inducers, their molecular mechanisms for triggering ICD initiation and subsequent protective antitumour immune responses, along with considerations for validating ICD both in vitro and in vivo. We also aim to offer insights into the future development of metal complexes with enhanced ICD-inducing properties and their applications in potentiating antitumour immunity.
Collapse
Affiliation(s)
- Jiao Xia Zou
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Meng Rui Chang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Nikita A Kuznetsov
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Jia Xuan Kee
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
- NUS Graduate School - Integrative Science and Engineering Programme (ISEP), National University of Singapore 21 Lower Kent Ridge Rd Singapore 119077 Singapore
| |
Collapse
|
13
|
Guo Y, Jiang T, Liang S, Wang A, Li J, Jia Y, Li Q, Yin J, Bai S, Li J. Immunostimulatory Hydrogel with Synergistic Blockage of Glutamine Metabolism and Chemodynamic Therapy for Postoperative Management of Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412507. [PMID: 39976234 PMCID: PMC12005773 DOI: 10.1002/advs.202412507] [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: 10/07/2024] [Revised: 01/13/2025] [Indexed: 02/21/2025]
Abstract
Glioblastoma multiforme (GBM) is one of the most lethal malignant brain tumors in the central nervous system. Patients face many challenges after surgery, including tumor recurrence, intracranial pressure increase due to cavitation, and limitations associated with immediate postoperative oral chemotherapy. Here an injected peptide gel with in situ immunostimulatory functions is developed to coordinate the regulation of glutamine metabolism and chemodynamic therapy for overcoming these postoperative obstacles. The methodology entails crafting injectable gel scaffolds with short peptide molecules, incorporating the glutaminase inhibitor CB-839 and copper peptide self-assembled particles (Cu-His NPs) renowned for their chemodynamic therapy (CDT) efficacy. By fine-tuning glutamic acid production via metabolic pathways, this system not only heightens the therapeutic prowess of copper peptide particles in CDT but also escalates intracellular oxidative stress. This dual mechanism culminates in augmented immunogenic cell death within glioblastoma multiforme cells and improves a conducive immune microenvironment. Based on the concept of metabolic reprogramming, this treatment strategy has great potential to significantly reduce GBM tumor recurrence and prolong median survival in murine models.
Collapse
Affiliation(s)
- Yiran Guo
- State Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijing100190China
- Key Laboratory of Carbohydrate Chemistry and BiotechnologyMinistry of EducationSchool of BiotechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Tianhe Jiang
- State Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijing100190China
| | - Sen Liang
- State Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijing100190China
| | - Anhe Wang
- State Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jieling Li
- State Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yi Jia
- University of Chinese Academy of SciencesBeijing100049China
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
| | - Qi Li
- State Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jian Yin
- Key Laboratory of Carbohydrate Chemistry and BiotechnologyMinistry of EducationSchool of BiotechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Shuo Bai
- State Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Junbai Li
- University of Chinese Academy of SciencesBeijing100049China
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
| |
Collapse
|
14
|
Zhang J, Tian T, Tian S, Yao J, Zhang Y, Xie R, Yang T, Han B. Study on the Mechanism of QRICH1 Mediating PRMT1 to Regulate the Arginine Methylation Modification of cGAS to Promote Arsenics-Induced Pyroptosis in Hepatocellular Carcinoma Cells. J Hepatocell Carcinoma 2025; 12:597-614. [PMID: 40124968 PMCID: PMC11930257 DOI: 10.2147/jhc.s505266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose This study aims to investigate the mechanism of action of arsenic-based agents against hepatocellular carcinoma (HCC) and to identify effective drug targets for HCC treatment. Methods Huh7 and HepG2 cells treated with NaAsO2 were assessed for cell viability, pyroptosis, migration, and invasion after undergoing lentiviral transfection. An orthotopic liver tumor model was established and divided into a model group and a treatment group. Proteins associated with QRICH1, PRMT1, cGAS-STING, and the classical pyroptosis pathway were quantified using Western blotting. The intracellular expression and localization of PRMT1 and NLRP3 in HCC were analyzed through cellular immunofluorescence. Co-immunoprecipitation (Co-IP) was performed to examine the protein interactions between PRMT1 and cGAS, as well as between STING and NLRP3. Chromatin immunoprecipitation (ChIP) was used to confirm QRICH1 enrichment in the PRMT1 promoter region. Results NaAsO2 treatment significantly inhibited the proliferation of Huh7 and HepG2 cells and effectively blocked their migration and invasion capabilities, while promoting cellular pyroptosis. Quantitative polymerase chain reaction(QRCR) and ChIP assays confirmed that NaAsO2 regulates PRMT1 expression by down-regulate QRICH1 binding in the PRMT1 promoter region. Additionally, NaAsO2 decreased the expression of the QRICH1-PRMT1 complex and upregulated the cGAS-STING signaling pathway, activating the downstream NLRP3-dependent classical pyroptosis pathway. Overexpression of QRICH1 reversed these effects. Conclusion NaAsO2 inhibits the expression of the QRICH1-PRMT1 axis, activates cGAS-STING signaling pathway transduction, and induces pyroptosis in HCC cells, thereby increasing the infiltration of immune cells in liver cancer tissues.
Collapse
Affiliation(s)
- Jiayuan Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Tian Tian
- Department of Eugenic Genetics, Guiyang Maternal and Child Health Care Hospital, Guiyang, Guizhou, 550003, People’s Republic of China
| | - Shanshan Tian
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Jinhai Yao
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Yingwan Zhang
- Qianxinan People’s Hospital, Qianxinan Affiliated Hospital of Zunyi Medical University, Xingyi, Guizhou, 562400, People’s Republic of China
| | - Rujia Xie
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Ting Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Bing Han
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| |
Collapse
|
15
|
Donati C, Hashim II, Pozsoni NB, Bourda L, Van Hecke K, Cazin CSJ, Visentin F, Nolan SP, Gandin V, Scattolin T. Investigation of the in vitro anticancer potential of bis(imino)acenaphthene-N-heterocyclic carbene transition metal complexes revealed TrxR inhibition and triggering of immunogenic cell death (ICD) for allyl palladates. RSC Med Chem 2025:d5md00039d. [PMID: 40171235 PMCID: PMC11956031 DOI: 10.1039/d5md00039d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/16/2025] [Indexed: 04/03/2025] Open
Abstract
Immunogenic cell death (ICD) is a regulated form of cell death that activates an immune response through the release of danger-associated molecular patterns (DAMPs), including calreticulin, ATP, and HMGB1. Gold complexes are known to induce ICD, but the ICD-inducing potential of palladium complexes remains largely unexplored. We report the first examples of palladium compounds capable of inducing ICD, specifically allyl palladates bearing bis(imino)acenaphthene-NHC (BIAN-NHC) ligands. Cytotoxicity tests on human cancer cell lines revealed that allyl palladates outperform their cinnamyl analogues and gold(i)/copper(i) BIAN-NHC complexes. Notably, [BIAN-IMes·H][PdCl2(allyl)] 2a showed excellent TrxR inhibition, reducing activity by 67% and surpassing auranofin. This inhibition strongly correlates with ICD induction, as evidenced by enhanced DAMP marker expression, including superior ATP and HMGB1 release compared to doxorubicin. These findings establish allyl palladates as a novel class of ICD inducers with dual anticancer activity and immune activation potential.
Collapse
Affiliation(s)
- Chiara Donati
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova via Marzolo 5 35131 Padova Italy
| | - Ishfaq Ibni Hashim
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University Krijgslaan 281,S-3 9000 Ghent Belgium
| | - Nestor Bracho Pozsoni
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University Krijgslaan 281,S-3 9000 Ghent Belgium
| | - Laurens Bourda
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University Krijgslaan 281,S-3 9000 Ghent Belgium
| | - Kristof Van Hecke
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University Krijgslaan 281,S-3 9000 Ghent Belgium
| | - Catherine S J Cazin
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University Krijgslaan 281,S-3 9000 Ghent Belgium
| | - Fabiano Visentin
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155 30174 Venezia-Mestre Italy
| | - Steven P Nolan
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University Krijgslaan 281,S-3 9000 Ghent Belgium
| | - Valentina Gandin
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova via Marzolo 5 35131 Padova Italy
| | - Thomas Scattolin
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova via Marzolo 1 35131 Padova Italy
| |
Collapse
|
16
|
Zhang Z, Xu C, Gong N, Qing G, Zhang Y, Shi Y, Brenner JS, Li F, Xu FJ, Liang XJ. An antigen-capturing and lymph node-targeting nanoparticle for cancer immunotherapy. J Control Release 2025; 379:993-1005. [PMID: 39889883 DOI: 10.1016/j.jconrel.2025.01.087] [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: 10/14/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Cancer immunotherapy leverages the immune system to combat cancer and has shown promise for many patients. However, its effectiveness is often hampered by an immunosuppressive tumor microenvironment and the low immunogenicity of tumor cells. In this study, we developed an in situ cancer vaccine that integrates chemotherapy and immunotherapy in a single platform. We synthesized two amphiphilic polymers with poly-albumin-binding domains (PABD) that can target the lymph nodes, PABD-PGEA and PABD-PGED. Compared with previous albumin-hijacking strategies utilizing the same albumin-binding domains, PABD-PGEA exhibited approximately six times greater lymph node-targeting ability, demonstrating enhanced antigen-capturing capability. We loaded PABD-PGEA with doxorubicin (DOX), a drug known to induce immunogenic cell death (ICD) in tumor cells, to form DOX@PABD-PGEA nanomicelles. DOX@PABD-PGEA inhibited tumor growth and extended the survival of mice with B16F10 melanoma through chemotherapy and immunotherapy. Notably, DOX@PABD-PGEA prevented tumor recurrence post-surgery by promoting efficient antigen presentation and reversing immunosuppression in the tumor microenvironment. Our findings suggest that DOX@PABD-PGEA, as an antigen-capturing nanoparticle, provides a safe and effective platform for in situ cancer vaccines and improves cancer immunotherapy.
Collapse
Affiliation(s)
- Zhen Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China
| | - Chen Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ningqiang Gong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China
| | - Guangchao Qing
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China
| | - Yuxuan Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China
| | - Yuxuan Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fangzhou Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| | - Fu-Jian Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
17
|
Alkayyal AA, Mahmoud AB. A 5-Year Update on the Clinical Development of Cancer Cell-Based Vaccines for Glioblastoma Multiforme. Pharmaceuticals (Basel) 2025; 18:376. [PMID: 40143152 PMCID: PMC11946125 DOI: 10.3390/ph18030376] [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: 11/04/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Glioblastoma multiforme (GBM) is considered one of the most aggressive forms of brain cancer with a 15-month median survival, despite advancements in surgery, radiotherapy, and chemotherapy. The immune-suppressed tumor microenvironment and the blood-brain barrier are major contributors to its poor prognosis and treatment resistance. In the last decade, significant progress has been made in developing cell-based vaccines to boost immune responses against GBM. This review provides an extensive update on recent clinical trials involving various cancer cell vaccines, including ICT-107, the α-type-1 DC vaccine, and others. Although these trials have demonstrated potential improvements in progression-free survival (PFS) and overall survival (OS), the diverse and immune-suppressed nature of GBM poses challenges for consistent therapeutic success. We discuss the details of these trials along with the potential mechanism of vaccine efficacy and immune activations. The findings of these trials highlight the significance of a personalized immunotherapy approach and suggest that patient stratification could significantly advance the clinical management of GBM.
Collapse
Affiliation(s)
- Almohanad A. Alkayyal
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Madinah 41477, Saudi Arabia
- Health and Life Research Center, Taibah University, Madinah 42353, Saudi Arabia
| |
Collapse
|
18
|
Zhang J, Wang F, Sun Z, Ye J, Chu H. Multidimensional applications of prussian blue-based nanoparticles in cancer immunotherapy. J Nanobiotechnology 2025; 23:161. [PMID: 40033359 PMCID: PMC11874808 DOI: 10.1186/s12951-025-03236-x] [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: 10/25/2024] [Accepted: 02/16/2025] [Indexed: 03/05/2025] Open
Abstract
Immunotherapy holds notable progress in the treatment of cancer. However, the clinical therapeutic effect remains a significant challenge due to immune-related side effects, poor immunogenicity, and immunosuppressive microenvironment. Nanoparticles have emerged as a revolutionary tool to surmount these obstacles and amplify the potency of immunotherapeutic agents. Prussian blue nanoparticles (PBNPs) exhibit multi-dimensional immune function in cancer immunotherapy, including acting as a nanocarrier to deliver immunotherapeutic agents, as a photothermal agent to improve the efficacy of immunotherapy through photothermal therapy, as a nanozyme to regulate tumor microenvironment, and as an iron donor to induce immune events related to ferroptosis and tumor-associated macrophages polarization. This review focuses on the advances and applications of PBNPs in cancer immunotherapy. First, the biomedical functions of PBNPs are introduced. Then, based on the immune function of PBNPs, we systematically reviewed the multidimensional application of PBNPs in cancer immunotherapy. Finally, the challenges and future developments of PBNPs-based cancer immunotherapy are highlighted.
Collapse
Affiliation(s)
- Jiayi Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Fang Wang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
19
|
Zhou L, Fan S, Zhang W, Gong Z, Wang D, Tang D. The battle within: cell death by phagocytosis in cancer. Clin Transl Oncol 2025; 27:871-886. [PMID: 39167272 DOI: 10.1007/s12094-024-03650-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
The process by which living cells are phagocytosed and digested to death is called cell death by phagocytosis, a term that has just recently been generalized and redefined. It is characterized by the phagocytosis of living cells and the cessation of cell death by phagocytosis. Phagocytosis of dead cells is a widely discussed issue in cancer, cell death by phagocytosis can stimulate phagocytosis and stimulate adaptive immunity in tumors, and at the same time, do not-eat-me signaling is an important site for cancer cells to evade recognition by phagocytes. Therefore, we discuss in this review cell death by phagocytosis occurring in cancer tissues and emphasize the difference between this new concept and the phagocytosis of dead tumor cells. Immediately thereafter, we describe the mechanisms by which cell death by phagocytosis occurs and how tumors escape phagocytosis. Finally, we summarize the potential clinical uses of cell death by phagocytosis in tumor therapy and strive to provide ideas for tumor therapy.
Collapse
Affiliation(s)
- Lujia Zhou
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Shiying Fan
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Wenjie Zhang
- School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Zhiyuan Gong
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
| |
Collapse
|
20
|
Wang X, Allen C. Synergistic effects of thermosensitive liposomal doxorubicin, mild hyperthermia, and radiotherapy in breast cancer management: an orthotopic mouse model study. Drug Deliv Transl Res 2025; 15:1011-1022. [PMID: 38977541 DOI: 10.1007/s13346-024-01654-2] [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: 06/13/2024] [Indexed: 07/10/2024]
Abstract
Liposome formulations of the cancer drug doxorubicin have been developed to address the severe side effects that result from administration of this drug in a conventional formulation. Among them, thermosensitive liposomal doxorubicin presents enhanced tumor targeting and efficient drug release when combined with mild hyperthermia localized to the tumor site. Exploiting the radiosensitizing benefits of localized thermal therapy, the integration of radiation therapy with the thermally activated liposomal system is posited to amplify the anti-tumor efficacy. This study explored a synergistic therapeutic strategy that combines thermosensitive liposomal doxorubicin, mild hyperthermia, and radiotherapy, using an orthotopic murine model of breast cancer. The protocol of sequential multi-modal treatment, incorporating low-dose chemotherapy and radiotherapy, substantially postponed the progression of primary tumor growth in comparison to the application of monotherapy at elevated dosages. Improvements in unheated distant lesions were also observed. Furthermore, the toxicity associated with the combination treatment was comparable to that of either thermosensitive liposome treatment or radiation alone at low doses. These outcomes underscore the potential of multi-modal therapeutic strategies to refine treatment efficacy while concurrently diminishing adverse effects in the management of breast cancer, providing valuable insight for the future refinement of thermosensitive liposomal doxorubicin applications.
Collapse
Affiliation(s)
- Xuehan Wang
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada.
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
21
|
Wang Y, Liu C, Pang J, Li Z, Zhang J, Dong L. The Extra-Tumoral Vaccine Effects of Apoptotic Bodies in the Advancement of Cancer Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410503. [PMID: 39871756 PMCID: PMC11878267 DOI: 10.1002/smll.202410503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/16/2025] [Indexed: 01/29/2025]
Abstract
The induction of apoptosis in tumor cells is a common target for the development of anti-tumor therapies; however, these therapies still leave patients at increased risk of disease recurrence. For example, apoptotic tumor cells can promote tumor growth and immune evasion via the secretion of metabolites, apoptotic extracellular vesicles, and induction of pro-tumorigenic macrophages. This paradox of apoptosis induction and the pro-tumorigenic effects of tumor cell apoptosis has begged the question of whether apoptosis is a suitable cancer therapy, and led to further explorations into other immunogenic cell death-based approaches. However, these strategies still face multiple challenges, the most critical of which is the tumor microenvironment. Contrary to the promotion of immune tolerance mediated by apoptotic tumor cells, apoptotic bodies with enriched tumor-related antigens have demonstrated great immunogenic potential, as evidenced by their ability to initiate systemic T-cell immune responses. These characteristics indicate that apoptotic body-based therapies could be ideal "in situ" extra-tumoral tumor vaccine candidates for the treatment of cancers, and further address the current issues with apoptosis-based or immunotherapy treatments. Although not yet tested clinically, apoptotic body-based vaccines have the potential to better treatment strategies and patient outcomes in the future.
Collapse
Affiliation(s)
- Yulian Wang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Chunyan Liu
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Jiayun Pang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Zhenjiang Li
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
- Chemistry and Biomedicine Innovative CenterNanjing UniversityNanjingJiangsu210023China
| |
Collapse
|
22
|
Wu YL, Zhao RR, Wu X, Liu CL, Liu CZ. Carrier-free nanodrug based on small molecule drug and sonosensitizer for enhanced the ferroptosis-driven multimodal synergistic therapy of prostate cancer. Eur J Pharm Biopharm 2025; 208:114656. [PMID: 39909320 DOI: 10.1016/j.ejpb.2025.114656] [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: 12/08/2024] [Revised: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Ferroptosis plays a significant role in overcoming the therapeutic resistance of cancer cells. Herein, a carrier-free nanoparticle based on small molecule drug sorafenib (SRF) and sonosensitizer rose bengal (RB) was constructed (named SR NPs) for ferroptosis-driven chemotherapy and sonodynamic synergistic therapy (SDT) of prostate cancer (PCa). SR NPs could be enriched in tumor cells and efficiently inhibit tumor cell proliferation. These nanodrugs could significantly reduce glutathione (GSH) synthesis, and inhibit glutathione peroxidase 4 (GPX4) expression by inhibiting the glutamate/cysteine antiporter system (System Xc-) pathway of ferroptosis. Moreover, SR NPs-mediated ferroptosis significantly improved the amount of reactive oxygen species (ROS) generated by SDT, inhibited cell migration and adhesion, enhanced the accumulation of lipid peroxides (LPO) and augmented chemo-sonodynamic therapy. Notably, in vivo studies demonstrated that SR NPs enhanced tumor accumulation, exhibited good biocompatibility, and showed high anti-tumor efficacy in PC-3 tumor-bearing mice. This work offered a new strategy to enhance the treatment efficacy of prostate cancer (PCa) through ferroptosis-chemotherapy-sonodynamic synergistic therapy.
Collapse
Affiliation(s)
- Yao-Lin Wu
- State Key Laboratory of Bio-fibers and Eco-Textiles, Institute of Biochemical Engineering, Affiliated Qingdao Central Hospital, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Rui-Rui Zhao
- State Key Laboratory of Bio-fibers and Eco-Textiles, Institute of Biochemical Engineering, Affiliated Qingdao Central Hospital, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China.
| | - Xiao Wu
- State Key Laboratory of Bio-fibers and Eco-Textiles, Institute of Biochemical Engineering, Affiliated Qingdao Central Hospital, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Chun-Lei Liu
- State Key Laboratory of Bio-fibers and Eco-Textiles, Institute of Biochemical Engineering, Affiliated Qingdao Central Hospital, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Chun-Zhao Liu
- State Key Laboratory of Bio-fibers and Eco-Textiles, Institute of Biochemical Engineering, Affiliated Qingdao Central Hospital, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
23
|
Cereghetti AS, Turko P, Cheng P, Benke S, Al Hrout A, Dzung A, Dummer R, Hottiger MO, Chahwan R, Ferretti LP, Levesque MP. DNA Methyltransferase Inhibition Upregulates the Costimulatory Molecule ICAM-1 and the Immunogenic Phenotype of Melanoma Cells. JID INNOVATIONS 2025; 5:100319. [PMID: 39867570 PMCID: PMC11759630 DOI: 10.1016/j.xjidi.2024.100319] [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: 01/26/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 01/28/2025] Open
Abstract
In cutaneous melanoma, epigenetic dysregulation is implicated in drug resistance and tumor immune escape. However, the epigenetic mechanisms that influence immune escape remain poorly understood. To elucidate how epigenetic dysregulation alters the expression of surface proteins that may be involved in drug targeting and immune escape, we performed a 3-dimensional surfaceome screen in primary melanoma cultures and identified the DNA-methyltransferase inhibitor decitabine as significantly upregulating the costimulatory molecule ICAM-1. By analyzing The Cancer Genome Atlas melanoma dataset, we further propose ICAM-1 upregulation on melanoma cells as a biomarker of a proinflammatory and antitumorigenic signature. Specifically, we showed that DNA-methyltransferase inhibitor administration upregulated the expression of the antigen-presenting machinery, HLA class I/II, as well as the secretion of the proinflammatory chemokines CXCL9 and CXCL10. Our in silico analysis on The Cancer Genome Atlas and ex vivo experiments on human primary melanoma samples revealed that increased ICAM-1 expression positively correlated with increased immunogenicity of human melanoma cells and correlated with increased immune cell infiltration. These findings suggest a therapeutic approach to modulate the immunogenic phenotype of melanoma cells, hence supporting the exploration of DNA-methyltransferase inhibitor as a potential inducer of infiltration in immunologically cold tumors.
Collapse
Affiliation(s)
| | - Patrick Turko
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Schlieren, Switzerland
| | - Phil Cheng
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Schlieren, Switzerland
| | - Stephan Benke
- Flow Cytometry Facility, University of Zurich, Zurich, Switzerland
| | - Ala’a Al Hrout
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Andreas Dzung
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Schlieren, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Schlieren, Switzerland
| | - Michael O. Hottiger
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Richard Chahwan
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Lorenza P. Ferretti
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Schlieren, Switzerland
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Mitchell P. Levesque
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Schlieren, Switzerland
| |
Collapse
|
24
|
Chen SY, Shyu IL, Chi JT. NINJ1 in Cell Death and Ferroptosis: Implications for Tumor Invasion and Metastasis. Cancers (Basel) 2025; 17:800. [PMID: 40075648 PMCID: PMC11898531 DOI: 10.3390/cancers17050800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
NINJ1 was initially recognized for its role in nerve regeneration and cellular adhesion. Subsequent studies have uncovered its participation in cancer progression, where NINJ1 regulates critical steps in tumor metastasis, such as cell migration and invasion. More recently, NINJ1 has emerged as a multifunctional protein mediating plasma membrane rupture (PMR) in several lytic cell death processes, including apoptosis, necroptosis, and pyroptosis. However, its role in ferroptosis-an iron-dependent form of lytic cell death characterized by lipid peroxidation-remained unclear until 2024. Ferroptosis is a tumor suppression mechanism that may be particularly relevant to detached and metastatic cancer cells. This review explores the role of NINJ1 in tumor invasion and metastasis, focusing on its regulation of ferroptosis via a non-canonical mechanism distinct from other cell deaths. We discuss the process of ferroptosis and its implications for cancer invasion and metastasis. Furthermore, we review recent studies highlighting the diverse roles of NINJ1 in ferroptosis regulation, including its canonical function in PMR and its non-canonical function of modulating intracellular levels of glutathione (GSH) and coenzyme A (CoA) via interaction with xCT anti-porter. Given that ferroptosis has been associated with tumor suppression, metastasis, the elimination of treatment-resistant cancer cells, and tumor dormancy, NINJ1's modulation of ferroptosis presents a promising therapeutic target for inhibiting metastasis. Understanding the dual role of NINJ1 in promoting or restraining ferroptosis depending on cellular context could open avenues for novel anti-cancer strategies to enhance ferroptotic vulnerability in metastatic tumors.
Collapse
Affiliation(s)
- Ssu-Yu Chen
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA;
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ing-Luen Shyu
- Department of Obstetrics and Gynecology, Chi Mei Medical Center, Tainan 710, Taiwan
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Center for Advanced Genomic Technologies, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
25
|
Sun F, Sun Y, Tian H. An Immunogenic Cell Death-Related Gene Signature Predicts the Prognosis and Immune Infiltration of Cervical Cancer. Cancer Inform 2025; 24:11769351251323239. [PMID: 40008391 PMCID: PMC11851768 DOI: 10.1177/11769351251323239] [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: 10/10/2024] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Objectives Immunogenic cell death (ICD) has been demonstrated to play a critical role in the development and progression of malignant tumors by modulating the anti-tumor immune response. However, its function in cervical cancer (CC) remains largely unexplored. In this study, we aimed to construct an ICD-related gene signature to predict patient prognosis and immune cell infiltration in CC. Methods The gene expression profiles and clinical data of CC were downloaded from The Cancer Genome Alas (TCGA) and Gene Expression Omnibus (GEO) datasets, serving as the training and testing groups, respectively. An ICD-related gene signature was developed using the LASSO-Cox model. The expression levels of the associated ICD-related genes were evaluated using single-cell data, CC cell lines, and clinical samples in vitro. Results Two ICD-associated subtypes (cluster 1 and cluster 2) were identified through consensus clustering. Patients classified into cluster 2 demonstrated higher levels of immune cell infiltration and exhibited a more favorable prognosis. Subsequently, an ICD-related gene signature comprising 3 genes (IL1B, IFNG, and FOXP3) was established for CC. Based on the median risk score, patients in both training and testing cohorts were segregated into high-risk and low-risk groups. Further analyses indicated that the estimated risk score functioned as an independent prognostic factor for CC and influenced immune cell abundance within the tumor microenvironment. The up-regulation of the identified ICD-related genes was further validated in CC cell lines and collected clinical samples. Conclusion In summary, the stratification based on ICD-related genes demonstrated strong efficacy in predicting patient prognosis and immune cell infiltration, which also provides valuable new perspectives for the diagnosis and prognosis of CC.
Collapse
Affiliation(s)
- Fangfang Sun
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, P. R. China
| | - Yuanyuan Sun
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, P. R. China
| | - Hui Tian
- School of Cyber Science and Engineering, Zhengzhou University, Zhengzhou, Henan Province, P. R. China
| |
Collapse
|
26
|
Li SR, Tao SY, Li Q, Hu CY, Sun ZJ. Harnessing nanomaterials for copper-induced cell death. Biomaterials 2025; 313:122805. [PMID: 39250865 DOI: 10.1016/j.biomaterials.2024.122805] [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: 03/12/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/11/2024]
Abstract
Copper (Cu), an essential micronutrient with redox properties, plays a pivotal role in a wide array of pathological and physiological processes across virtually all cell types. Maintaining an optimal copper concentration is critical for cellular survival: insufficient copper levels disrupt respiration and metabolism, while excess copper compromises cell viability, potentially leading to cell death. Similarly, in the context of cancer, copper exhibits a dual role: appropriate amount of copper can promote tumor progression and be an accomplice, yet beyond befitting level, copper can bring about multiple types of cell death, including autophagy, apoptosis, ferroptosis, immunogenic cell death, pyroptosis, and cuproptosis. These forms of cell death are beneficial against cancer progression; however, achieving precise copper regulation within tumors remains a significant challenge in the pursuit of effective cancer therapies. The emergence of nanodrug delivery systems, distinguished by their precise targeting, controlled release, high payload capacity, and the ability to co-deliver multiple agents, has revitalized interest in exploiting copper's precise regulatory capabilities. Nevertheless, there remains a dearth of comprehensive review of copper's bidirectional effects on tumorigenesis and the role of copper-based nanomaterials in modulating tumor progression. This paper aims to address this gap by elucidating the complex role in cancer biology and highlighting its potential as a therapeutic target. Through an exploration of copper's dualistic nature and the application of nanotechnology, this review seeks to offer novel insights and guide future research in advancing cancer treatment.
Collapse
Affiliation(s)
- Su-Ran Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China
| | - Shi-Yue Tao
- Bathune School of Stomatology, Jilin University, Changchun, 130021, Jilin, PR China
| | - Qian Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China
| | - Chuan-Yu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China.
| |
Collapse
|
27
|
Sebastião AI, Simões G, Oliveira F, Mateus D, Falcão A, Carrascal MA, Gomes C, Neves B, Cruz MT. Dendritic cells in triple-negative breast cancer: From pathophysiology to therapeutic applications. Cancer Treat Rev 2025; 133:102884. [PMID: 39837068 DOI: 10.1016/j.ctrv.2025.102884] [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: 09/21/2024] [Revised: 11/28/2024] [Accepted: 01/11/2025] [Indexed: 01/23/2025]
Abstract
Breast cancer is the second most commonly diagnosed cancer in women and the fifth leading cause of cancer-related deaths worldwide. It is a highly heterogeneous disease, consisting of multiple subtypes that vary significantly in clinical characteristics and survival outcomes. Triple-negative breast cancer (TNBC) is a particularly aggressive and challenging subtype of breast cancer. Several immunotherapeutic approaches have been tested in patients with TNBC to improve disease outcomes, including the administration of dendritic cell (DC)-based vaccines. DCs are a heterogeneous cell population that play a crucial role in bridging the innate and adaptive immune systems. Therefore, DCs have been increasingly used in cancer vaccines due to their ability to prime and boost antigen specific T-cell immune responses. This review aims to provide a comprehensive overview of TNBC, including potential targets and pharmacological strategies, as well as an overview of DCs and their relevance in TNBC. In addition, we review ongoing clinical trials and shed light on the evolving landscape of DC-based immunotherapy for TNBC.
Collapse
Affiliation(s)
- Ana Isabel Sebastião
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Gonçalo Simões
- Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Filomena Oliveira
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Daniela Mateus
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; BioMark@UC/CEB-LABBELS, Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
| | | | - Célia Gomes
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research - iCBR, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Bruno Neves
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal; Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|
28
|
Yang T, Dai L, Liu J, Lu Y, Pan M, Pan L, Ye L, Yuan L, Li X, Bei Z, Qian Z. Metal-phenolic-network-coated gold nanoclusters for enhanced photothermal/chemodynamic/immunogenic cancer therapy. Bioact Mater 2025; 44:447-460. [PMID: 39534788 PMCID: PMC11555603 DOI: 10.1016/j.bioactmat.2024.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/03/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, characterised by a short survival period, high malignancy, strong invasiveness, and high rates of recurrence and metastasis. Due to its unique molecular phenotype, TNBC is insensitive to endocrine therapy or molecular targeted therapy. The conventional treatment approach involves systemic chemotherapy for overall management; however, adjuvant chemotherapy after surgery has shown poor efficacy as residual lesions can easily lead to tumour recurrence. Therefore, there is an urgent need to find more effective treatment strategies. Herein, we designed a gold nanocluster coated with a metal-phenol formaldehyde network structure (AuNCs@PDA-Mn) for tumour Photothermal therapy and chemodynamic therapy (PTT and CDT), which induces systemic immune responses to suppress tumour metastasis. Experimental results show that after continuous irradiation for 10 min under an 808 nm laser (1.0W/cm2), AuNCs@PDA-Mn not only exhibits better tumour inhibition both in vitro and in vivo but also triggers stronger immune effects systemically. Therefore, this combined PTT and CDT treatment approach has great potential and provides a clinically relevant and valuable option for triple-negative breast cancer.
Collapse
Affiliation(s)
- Tingyu Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liqun Dai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi Lu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Pan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lili Pan
- Department of Nuclear Medicine and Clinical Nuclear Medicine Research Lab, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lin Ye
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Liping Yuan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xicheng Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhongwu Bei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
29
|
Khilar S, Dembinska-Kenner A, Hall H, Syrmos N, Ligarotti GKI, Plaha P, Apostolopoulos V, Chibbaro S, Barbagallo GMV, Ganau M. Towards a New Dawn for Neuro-Oncology: Nanomedicine at the Service of Drug Delivery for Primary and Secondary Brain Tumours. Brain Sci 2025; 15:136. [PMID: 40002469 PMCID: PMC11852924 DOI: 10.3390/brainsci15020136] [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: 12/12/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
(1) Background/Objectives: Primary and secondary brain tumours often hold devastating prognoses and low survival rates despite the application of maximal neurosurgical resection, and state-of-the-art radiotherapy and chemotherapy. One limiting factor in their management is that several antineoplastic agents are unable to cross the blood-brain barrier (BBB) to reach the tumour microenvironment. Nanomedicine could hold the potential to become an effective means of drug delivery to overcome previous hurdles towards effective neuro-oncological treatments. (2) Methods: A scoping review following the PRISMA-ScR guidelines and checklist was conducted using key terms input into PubMed to find articles that reflect emerging trends in the utilisation of nanomedicine in drug delivery for primary and secondary brain tumours. (3) Results: The review highlights various strategies by which different nanoparticles can be exploited to bypass the BBB; we provide a synthesis of the literature on the ongoing contributions to therapeutic protocols based on chemotherapy, immunotherapy, focused ultrasound, radiotherapy/radiosurgery, and radio-immunotherapy. (4) Conclusions: The emerging trends summarised in this scoping review indicate encouraging advantageous properties of nanoparticles as potential effective drug delivery mechanisms; however, there are still nanotoxicity issues that largely remain to be addressed before the translation of these innovations from laboratory to clinical practice.
Collapse
Affiliation(s)
- Smita Khilar
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
| | - Antonina Dembinska-Kenner
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
| | - Helen Hall
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
| | - Nikolaos Syrmos
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | | - Puneet Plaha
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
| | - Vasileios Apostolopoulos
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
| | - Salvatore Chibbaro
- Neurosurgery Unit, Department of Medical and Surgical Sciences and Neurosciences, Siena University, 53100 Siena, Italy
| | | | - Mario Ganau
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
30
|
Hou G, Chen Y, Lei H, Lu S, Cheng L. Nanomaterials-Induced PANoptosis: A Promising Anti-Tumor Strategy. Angew Chem Int Ed Engl 2025; 64:e202419649. [PMID: 39560000 DOI: 10.1002/anie.202419649] [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: 10/11/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/20/2024]
Abstract
Malignant tumors pose a significant threat to global public health. Promoting programmed cell death in cancer cells has become a critical strategy for cancer treatment. PANoptosis, a newly discovered form of regulated cell death, integrates key molecular components of pyroptosis, apoptosis, and necroptosis, activating these three death pathways simultaneously to achieve synergistic multi-mechanistic killing. PANoptosis significantly inhibits cancer cell growth and resistance and activates strong anti-tumor immune response, making tumor-specific induction of PANoptosis a potential cancer therapeutic strategy. Currently, cancer treatment research related to PANoptosis is focused mainly on the development of small molecules and cytokines. However, these approaches still face limitations in terms of metabolic stability and tumor specificity. The unique physicochemical properties and biological activities of nanomaterials hold significant promise for optimizing PANoptosis induction strategies. This review summarizes the concept and mechanisms of PANoptosis, highlights the latest applications of nanoagents in PANoptosis-based anti-cancer therapy, and discusses the challenges and future directions for clinical translation. It is hoped that this review will inspire further exploration and development of PANoptosis-based cancer treatments, providing new perspectives for researchers in the field.
Collapse
Affiliation(s)
- Guanghui Hou
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Youdong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Shunyi Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
31
|
Sun G, He L. A new paradigm for cancer immunotherapy: targeting immunogenic cell death-related noncoding RNA. Front Immunol 2025; 15:1498781. [PMID: 39916954 PMCID: PMC11798941 DOI: 10.3389/fimmu.2024.1498781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/30/2024] [Indexed: 02/09/2025] Open
Abstract
Cancer immunotherapy has shown significant potential in treating several malignancies by stimulating the host immune system to recognize and attack cancer cells. Immunogenic cell death (ICD) can amplify the antitumor immune responses and reverse the immunosuppressive tumor microenvironment, thus increasing the sensitivity of cancer immunotherapy. In recent years, noncoding RNAs (ncRNAs) have emerged as key regulatory factors in ICD and oncologic immunity. Accordingly, ICD-related ncRNAs hold promise as novel therapeutic targets for optimizing the efficacy of cancer immunotherapy. However, the immunomodulatory properties of ICD-related ncRNAs have not yet been comprehensively summarized. Hence, we summarize the current knowledge on ncRNAs involved in ICD and their potential roles in cancer immunotherapy in this review. It deepens our understanding of ncRNAs associated with ICD and provides a new strategy to enhance cancer immunotherapy by specifically targeting the ICD-related ncRNAs.
Collapse
Affiliation(s)
| | - Ling He
- The Ward Section of Home Overseas Doctors, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
32
|
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
|
33
|
Xue D, Xu M, Madden MD, Lian X, Older EA, Pulliam C, Hui Y, Shang Z, Gupta G, Raja MK, Wang Y, Sardi A, Long Y, Chen H, Fan D, Bugni TS, Testerman TL, Wu Q, Li J. Discovery of a Chimeric Polyketide Family as Cancer Immunogenic Chemotherapeutic Leads. J Am Chem Soc 2025; 147:265-277. [PMID: 39731542 DOI: 10.1021/jacs.4c09582] [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] [Indexed: 12/30/2024]
Abstract
Discovery of cancer immunogenic chemotherapeutics represents an emerging, highly promising direction for cancer treatment that uses a chemical drug to achieve the efficacy of both chemotherapy and immunotherapy. Herein, we report a high-throughput screening platform and the subsequent discovery of a new class of cancer immunogenic chemotherapeutic leads. Our platform integrates informatics-based activity metabolomics for the rapid identification of microbial natural products with both novel structures and potent activities. Additionally, we demonstrate the use of microcrystal electron diffraction (MicroED) for direct structure elucidation of lead compounds from partially purified mixtures. Using this strategy to screen geographically and phylogenetically diverse microbial metabolites against pseudomyxoma peritonei, a rare and severe cancer, we discovered a new class of leads, aspercyclicins. The aspercyclicins feature an unprecedented tightly packed polycyclic polyketide scaffold that comprises continuous fused, bridged, and spiro rings. The biogenesis of aspercyclicins involves two distinct biosynthetic pathways, leading to formation of chimeric compounds that cannot be predicted by bottom-up approaches mining natural product biosynthetic genes. With comparable potency to some clinically used anticancer drugs, aspercyclicins are active against multiple cancer cell types by inducing immunogenic cell death (ICD), including the release of damage-associated molecular patterns and subsequent phagocytosis of cancer cells. The broad-spectrum ICD-inducing activity of aspercyclicins, combined with their low toxicity to normal cells, represents a new class of potential cancer immunogenic chemotherapeutics and, particularly, the first drug lead for pseudomyxoma peritonei treatment.
Collapse
Affiliation(s)
- Dan Xue
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Mingming Xu
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Michael D Madden
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Xiaoying Lian
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Ethan A Older
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Conor Pulliam
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Yvonne Hui
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, United States
| | - Zhuo Shang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Gourab Gupta
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Manikanda K Raja
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, United States
| | - Armando Sardi
- Department of Surgical Oncology, The Institute for Cancer Care at Mercy, Mercy Medical Center, Baltimore, Maryland 21202, United States
| | - Yaoling Long
- Department of Biological and Physical Sciences, South Carolina State University, Orangeburg, South Carolina 29117, United States
| | - Hexin Chen
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, United States
| | - Tim S Bugni
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Lachman Institute for Pharmaceutical Development, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Traci L Testerman
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, United States
| | - Qihao Wu
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jie Li
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| |
Collapse
|
34
|
Christensen RL, Son HG, Zhou EY, Olesen UH, Garibyan L, Farinelli WA, Sakamoto FH, Rox Anderson R, Haedersdal M, Demehri S. Cutaneous Immune Responses to Ablative Fractional Laser, Heat- and Cold-Based Dermatological Procedures. Lasers Surg Med 2025; 57:101-111. [PMID: 39698750 DOI: 10.1002/lsm.23868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/24/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024]
Abstract
OBJECTIVE Physical treatment modalities, such as ablative fractional laser (AFL), electrocautery, and cryotherapy, are extensively used in the field of dermatology. This study aimed to characterize the short-term innate and adaptive immune responses induced by AFL compared with heat- and cold-based procedures. MATERIALS AND METHODS Innate (CD11b+Ly6G+ neutrophils) and adaptive (CD8+CD3+ T cells) immune cell infiltration and histopathological changes were examined in murine skin on Days 1 and 7, following AFL, monopolar-electrocautery (RF), thermocautery, and cryotherapy. Interventions were standardized to reach the reticular dermis. Clinical skin reactions were photo-documented daily. As a comparator, the adaptive immune response was examined in murine basal cell carcinomas (BCC) on Day 7 after AFL exposure. RESULTS Baseline histopathology confirmed immediate deep dermal tissue impact by all procedures. Immune cell dynamics varied across treatments throughout the progression of clinical and histopathological responses. On Day 1, AFL and heat-based procedures triggered an innate immune response, characterized by CD11b+Ly6G+ neutrophil cell infiltration that correlated with histopathological findings and immediate onset of clinical skin reactions. In addition, heat-based procedures led to an increase in overall dermal CD45+ cells (Day 1), which continued to rise for AFL and RF-electrocautery at Day 7 posttreatment. On the contrary, cryotherapy did not induce immediate (Day 1) innate immune responses, but instead a delayed increase in neutrophil and CD45+ cell infiltration (Day 7), which coincided with the late onset of clinical reaction. CD3+ T cells and CD8+CD3+ T cells demonstrated a similar pattern, with an increase observed for heat-based procedures on Day 1 and a delayed increase for cryotherapy on Day 7. Distinctive for AFL-treated skin, the level of dermal CD3+ T cells increased over time, significant by Day 7, and AFL-treated mouse BCCs responded with increased CD8+ T cell infiltration at Day 7 posttreatment. CONCLUSION Heat- and cold-based procedures developed distinct cutaneous immune responses, with cryotherapy resulting in a delayed response compared to immediate immune responses from heat-based procedures. The substantial T cell response induced by AFL in the skin and BCC tumors indicates a potential for AFL as an adjuvant in immunotherapeutic treatments of keratinocyte cancers.
Collapse
Affiliation(s)
- Rikke L Christensen
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Dermatology and Venereology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Heehwa G Son
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eray Yihui Zhou
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Uffe H Olesen
- Department of Dermatology and Venereology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Lilit Garibyan
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - William A Farinelli
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Fernanda H Sakamoto
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - R Rox Anderson
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Merete Haedersdal
- Department of Dermatology and Venereology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Ahmad I, Altameemi KKA, Hani MM, Ali AM, Shareef HK, Hassan ZF, Alubiady MHS, Al-Abdeen SHZ, Shakier HG, Redhee AH. Shifting cold to hot tumors by nanoparticle-loaded drugs and products. Clin Transl Oncol 2025; 27:42-69. [PMID: 38922537 DOI: 10.1007/s12094-024-03577-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
Cold tumors lack antitumor immunity and are resistant to therapy, representing a major challenge in cancer medicine. Because of the immunosuppressive spirit of the tumor microenvironment (TME), this form of tumor has a low response to immunotherapy, radiotherapy, and also chemotherapy. Cold tumors have low infiltration of immune cells and a high expression of co-inhibitory molecules, such as immune checkpoints and immunosuppressive molecules. Therefore, targeting TME and remodeling immunity in cold tumors can improve the chance of tumor repression after therapy. However, tumor stroma prevents the infiltration of inflammatory cells and hinders the penetration of diverse molecules and drugs. Nanoparticles are an intriguing tool for the delivery of immune modulatory agents and shifting cold to hot tumors. In this review article, we discuss the mechanisms underlying the ability of nanoparticles loaded with different drugs and products to modulate TME and enhance immune cell infiltration. We also focus on newest progresses in the design and development of nanoparticle-based strategies for changing cold to hot tumors. These include the use of nanoparticles for targeted delivery of immunomodulatory agents, such as cytokines, small molecules, and checkpoint inhibitors, and for co-delivery of chemotherapy drugs and immunomodulatory agents. Furthermore, we discuss the potential of nanoparticles for enhancing the efficacy of cancer vaccines and cell therapy for overcoming resistance to treatment.
Collapse
Affiliation(s)
- Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia.
| | | | - Mohaned Mohammed Hani
- Department of Medical Instrumentation Engineering Techniques, Imam Ja'afar Al-Sadiq University, Al Muthanna, Iraq
| | - Afaq Mahdi Ali
- Department of Pharmaceutics, Al-Turath University College, Baghdad, Iraq
| | - Hasanain Khaleel Shareef
- Department of Medical Biotechnology, College of Science, Al-Mustaqbal University, Hilla, Iraq
- Biology Department, College of Science for Women, University of Babylon, Hilla, Iraq
| | | | | | | | | | - Ahmed Huseen Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
36
|
Dadwal S, Dhar S, Baghel K, Mishra A, Mehrotra S, Prajapati VK. From past to present: The evolution of immunotherapy and its modern modalities. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 144:1-32. [PMID: 39978965 DOI: 10.1016/bs.apcsb.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Immunotherapy is emerging as a novel and reliable therapeutic technique for treating diseases such as autoimmunity, HIV/AIDS, allergy and cancers. This approach works by modulating the patient's immune system, activating both the innate and humoral branches to combat life-threatening diseases. The foundation of immunotherapy began with the discovery and development of "serum therapy" by German physiologist Emil Von Behring who received the Nobel Prize in 1901 for his contributions to the treatment of diphtheria. Around the same time, Dr. William Coley expanded the field for cancer treatment by developing the first immune based cure for sarcomas using attenuated strains of bacteria injected directly into patient's tumours. As medical science advanced, a broader understanding of the immune system and its components led to the emergence of different immunotherapeutic techniques. These include adoptive cell transfer therapy, cytokine therapy, cancer vaccines, and antibody-drug conjugates. The chapter provides a comprehensive understanding of the history and the current techniques used in immunotherapy, detailing the principles behind their mechanisms and the types of diseases tackled by each immunotherapeutic technique. By examining the journey from early discoveries to modern advancements, the chapter highlights the transformative impact of immunotherapy on medical science and patient care.
Collapse
Affiliation(s)
- Surbhi Dadwal
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sarthak Dhar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Kirti Baghel
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
37
|
Gao C, Zhang H, Wang X. Current advances on the role of ferroptosis in tumor immune evasion. Discov Oncol 2024; 15:736. [PMID: 39621177 PMCID: PMC11612115 DOI: 10.1007/s12672-024-01573-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/11/2024] [Indexed: 12/06/2024] Open
Abstract
Ferroptosis is a non-apoptotic form of regulated cell death characterized by iron accumulation and uncontrolled lipid peroxidation, leading to plasma membrane rupture and intracellular content release. Cancer immunotherapy, especially immune checkpoint inhibitors (ICIs) targeting PD-1 and PD-L1, has been considered a breakthrough in cancer treatment, achieving encouraging clinical anti-tumor effects in a variety of cancers. However, tumor immune evasion is indispensable to immunotherapy failure. The mechanisms of tumor immune evasion are quite complex, and its occurrence is inseparable from the ferroptosis in tumor microenvironment (TME). Thus, a comprehensive understanding of the role of ferroptosis in tumor immune evasion is crucial to enhance the efficacy of immunotherapy. In this review, we provide an overview of the recent advancements in understanding ferroptosis in cancer, covering molecular mechanisms and interactions with the TME. We also summarize the potential applications of ferroptosis induction in immunotherapy, as well as ferroptosis inhibition for cancer treatment in various conditions. We finally discuss ferroptosis as a double-edged sword, including the current challenges and future directions regarding its potential for cancer treatment.
Collapse
Affiliation(s)
- Changlin Gao
- Graduate School of Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Haoran Zhang
- Central Hospital Affiliated to Dalian University of Technology, Dalian, 116000, Liaoning, China
- Graduate School of Dalian Medical University, Dalian, 116000, Liaoning, China
| | - Xianwei Wang
- Central Hospital Affiliated to Dalian University of Technology, Dalian, 116000, Liaoning, China.
| |
Collapse
|
38
|
Brüßeler MT, Zam A, Moreno-Zafra VM, Rouatbi N, Hassuneh OWM, Marrocu A, Liam-Or R, Abdel-Bar HM, Walters AA, Al-Jamal KT. Polyinosinic/Polycytidylic Lipid Nanoparticles Enhance Immune Cell Infiltration and Improve Survival in the Glioblastoma Mouse Model. Mol Pharm 2024; 21:6339-6352. [PMID: 39556101 PMCID: PMC11615939 DOI: 10.1021/acs.molpharmaceut.4c00875] [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: 08/04/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024]
Abstract
Glioblastoma (GBM) immunotherapy is particularly challenging due to the pro-tumorigenic microenvironment, marked by low levels and inactive immune cells. Toll-like receptor (TLR) agonists have emerged as potent immune adjuvants but failed to show improved outcomes in clinical trials when administered as a monotherapy. We hypothesize that a combined nanoparticulate formulation of TLR agonist and immunogenic cell death-inducing drug (doxorubicin) will synergize to induce improved GBM immunotherapy. Lipid nanoparticle (LNP) formulations of the TLR agonists CpG and polyinosinic/polycytidylic (pIpC), with and without Dox, were first prepared, achieving an encapsulation efficiency >75% and a size <140 nm. In vitro studies identified that LNP pIpC was superior to CpG at activating bone marrow-derived immune cell populations (dendritic cells and macrophages) with minimal toxicity. It was also observed that the pIpC formulation can skew macrophage polarization toward the antitumorigenic M1 phenotype and increase macrophage phagocytosis of cancer cells. Upon intratumoral administration, pIpC Dox LNPs led to significant immune cell infiltration and activation. In survival models, the inclusion of Dox into pIpC LNP improved mice survival compared to control. However, addition of Dox did not show significant improvement in mice's survival compared to singly formulated pIpC LNP. This study has illustrated the potential of pIpC LNP formulations in prospective GBM immunotherapeutic regimes. Future studies will focus on optimizing dosage regimen and/or combination with other modalities, including the standard of care (temozolomide), immune checkpoint blockade, or cancer vaccines.
Collapse
Affiliation(s)
- Melanie
M. T. Brüßeler
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
- Ludwig
Maximilians University, Bayern, Munich, München 80539, Germany
| | - Alaa Zam
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Víctor M. Moreno-Zafra
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Nadia Rouatbi
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Osama W. M. Hassuneh
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Alessia Marrocu
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Revadee Liam-Or
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong 999077, China
| | - Hend Mohamed Abdel-Bar
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
- Department
of Pharmaceutics, Faculty of Pharmacy, University
of Sadat City, P.O. Box 32958, El Sadat, Egypt
| | - Adam Alexander Walters
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Khuloud T. Al-Jamal
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong 999077, China
| |
Collapse
|
39
|
Ren D, Xiong S, Ren Y, Yang X, Zhao X, Jin J, Xu M, Liang T, Guo L, Weng L. Advances in therapeutic cancer vaccines: Harnessing immune adjuvants for enhanced efficacy and future perspectives. Comput Struct Biotechnol J 2024; 23:1833-1843. [PMID: 38707540 PMCID: PMC11066472 DOI: 10.1016/j.csbj.2024.04.054] [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: 02/23/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/07/2024] Open
Abstract
Preventive cancer vaccines are highly effective in preventing viral infection-induced cancer, but advances in therapeutic cancer vaccines with a focus on eliminating cancer cells through immunotherapy are limited. To develop therapeutic cancer vaccines, the integration of optimal adjuvants is a potential strategy to enhance or complement existing therapeutic approaches. However, conventional adjuvants do not satisfy the criteria of clinical trials for therapeutic cancer vaccines. To improve the effects of adjuvants in therapeutic cancer vaccines, effective vaccination strategies must be formulated and novel adjuvants must be identified. This review offers an overview of the current advancements in therapeutic cancer vaccines and highlights in situ vaccination approaches that can be synergistically combined with other immunotherapies by harnessing the adjuvant effects. Additionally, the refinement of adjuvant systems using cutting-edge technologies and the elucidation of molecular mechanisms underlying immunogenic cell death to facilitate the development of innovative adjuvants have been discussed.
Collapse
Affiliation(s)
- Dekang Ren
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Shizheng Xiong
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Yujie Ren
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Xueni Yang
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Xinmiao Zhao
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Jiaming Jin
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Miaomiao Xu
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Li Guo
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lixing Weng
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| |
Collapse
|
40
|
Huang D, Yao Y, Lou Y, Kou L, Yao Q, Chen R. Disulfiram and cancer immunotherapy: Advanced nano-delivery systems and potential therapeutic strategies. Int J Pharm X 2024; 8:100307. [PMID: 39678262 PMCID: PMC11638648 DOI: 10.1016/j.ijpx.2024.100307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
The initial focus of the clinical application of disulfiram was its efficacy in treating alcoholism. However, recent research has revealed its potential as an anti-tumor agent and even as an enhancer of cancer immunotherapy. Disulfiram has received safety approval from the FDA, indicating its safety advantages over other substances used for disease treatment. Although clinical trials have been conducted on strategies involving disulfiram or its combination with other anti-tumor drugs, the treatment outcomes have not yielded satisfactory results, thereby emphasizing the significance of addressing drug delivery as a crucial challenge to be resolved. The need to explore advanced nano-delivery systems and the potential immunotherapy enhancement effect of disulfiram in cancer treatment has increased. This review highlights various ways in which disulfiram can combat cancer and importantly, activate immune-related mechanisms. It also discusses obstacles related to delivering disulfiram and provides existing solutions in terms of drug delivery. These drug delivery strategies offer solutions to address various challenges encountered in diverse delivery methods and aim to achieve enhanced therapeutic effects. The focus is on recent advancements in disulfiram delivery strategies and the future potential of disulfiram in immune regulation.
Collapse
Affiliation(s)
- Di Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Yinsha Yao
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Yifei Lou
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
41
|
Wu F, Xu Y. Immunogenic cell death-related cancer-associated fibroblast clusters and prognostic risk model in cervical cancer. APL Bioeng 2024; 8:046114. [PMID: 39691350 PMCID: PMC11650426 DOI: 10.1063/5.0240772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/01/2024] [Indexed: 12/19/2024] Open
Abstract
Cervical cancer (CC) remains a leading cause of female cancer mortality globally. Immunogenic cell death (ICD) influences the tumor microenvironment (TME) and adaptive immune responses. Cancer-associated fibroblasts (CAFs) within the TME suppress anti-tumor immunity and contribute to CC progression. This study identified three ICD-related CAF clusters linked to patient survival, including IL6+CAF and ILR1+CAF, which were associated with clinical outcomes. Using a nine-gene risk model, patients were stratified into risk groups, with high-risk individuals showing worse survival and correlations with pathways such as hypoxia and TGFβ. The model also predicted immunotherapy responses, highlighting immune infiltration differences across risk groups. These findings provide insights into the role of CAF clusters in CC and present a risk model that supports prognosis prediction and personalized therapy.
Collapse
Affiliation(s)
- Fei Wu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Yue Xu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| |
Collapse
|
42
|
Zhu Y, Li Y, Li X, Yu Y, Zhang L, Zhang H, Chen C, Chen D, Wang M, Xing N, Yang F, Wasilijiang W, Ye X. Targeting Hypoxia and Autophagy Inhibition via Delivering Sonodynamic Nanoparticles With HIF-2α Inhibitor for Enhancing Immunotherapy in Renal Cell Carcinoma. Adv Healthc Mater 2024; 13:e2402973. [PMID: 39396375 DOI: 10.1002/adhm.202402973] [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: 08/09/2024] [Revised: 09/28/2024] [Indexed: 10/15/2024]
Abstract
Immune checkpoint blockers (ICBs) therapy stands as the first-line treatment option for advanced renal cell carcinoma (RCC). However, its effectiveness is hindered by the immunosuppressive tumor microenvironment (TME). Sonodynamic therapy (SDT) generates tumor cell fragments that can prime the host's antitumor immunity. Nevertheless, the hypoxic microenvironment and upregulated autophagy following SDT often lead to cancer cell resistance. In response to these challenges, a hypoxia-responsive polymer (Poly(4,4'-azobisbenzenemethanol-PMDA)-mPEG5k, P-APm) encapsulating both a HIF-2α inhibitor (belzutifan) and the ultrasonic sensitize (Chlorin e6, Ce6) is designed, to create the nanoparticle APm/Ce6/HIF. APm/Ce6/HIF combined with ultrasound (US) significantly suppresses tumor growth and activates antitumor immunity in vivo. Moreover, this treatment effectively transforms the immunosuppressive microenvironment from "immune-cold" to "immune-hot", thereby enhancing the response to ICBs therapy. The findings indicate that APm/Ce6/HIF offers a synergistic approach combining targeted therapy with immunotherapy, providing new possibilities for treating RCC.
Collapse
Affiliation(s)
- Yihao Zhu
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yajian Li
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xuwen Li
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuan Yu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, 310022, China
| | - Lingpu Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hanchen Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Can Chen
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China
| | - Dong Chen
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Mingshuai Wang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Nianzeng Xing
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Feiya Yang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wahafu Wasilijiang
- Department of Urology, 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 Urology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Shanxi, 030013, China
| | - Xiongjun Ye
- Department of Urology, 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
|
43
|
Zhang X, Deng X, Tan J, Liu H, Zhang H, Li C, Li Q, Zhou J, Xiao Z, Li J. Idarubicin-loaded degradable hydrogel for TACE therapy enhances anti-tumor immunity in hepatocellular carcinoma. Mater Today Bio 2024; 29:101343. [PMID: 39687797 PMCID: PMC11647502 DOI: 10.1016/j.mtbio.2024.101343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/22/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a common and deadly cancer, often diagnosed at advanced stages, limiting surgical options. Transcatheter arterial chemoembolization (TACE) is a primary treatment for inoperable and involves the use of drug-eluting microspheres to slowly release chemotherapy drugs. However, patient responses to TACE vary, with some experiencing tumor progression and recurrence. Traditional TACE uses agents like oil-based drug emulsions and polyvinyl alcohol particles, which can permanently block blood vessels and increase tumor hypoxia. Additionally, TACE can suppress the immune system by reducing immune cell numbers and function, contributing to poor treatment outcomes. New approaches, like TACE using degradable starch microspheres and hydrogel-based materials, offer the potential to create different tumor environments that could improve both safety and efficacy. In our research, we developed a composite hydrogel (IF@Gel) made of Poloxamer-407 gel and Fe3O4 nanoparticles, loaded with idarubicin, to use as an embolic material for TACE in a rat model of orthotopic HCC. We observed promising therapeutic effects and investigated the impact on the tumor immune microenvironment, focusing on the role of immunogenic cell death (ICD). The composite hydrogel demonstrated excellent potential as an embolic material for TACE, and IF@Gel-based TACE demonstrated significant efficacy in rat HCC. Furthermore, our findings highlight the potential synergistic effects of ICD with anti-PD-L1 therapy, providing new insights into HCC treatment strategies. This study aims to provide improved treatment options for HCC and to deepen our understanding of the mechanisms of TACE and tumor environment regulation.
Collapse
Affiliation(s)
- Xiaokai Zhang
- Department of Hepatobiliopancreatic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450003, China
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiujiao Deng
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Jizhou Tan
- Department of Stomatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Haikuan Liu
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Hong Zhang
- Department of Interventional Radiology and Vascular Surgery, The Sixth Affiliated Hospital of Jinan University, Dongguan 523067, China
| | - Chengzhi Li
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Qingjun Li
- Department of Hepatobiliopancreatic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450003, China
| | - Jinxue Zhou
- Department of Hepatobiliopancreatic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450003, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Jiaping Li
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
44
|
Ji J, Ma Y, Liu X, Zhou Q, Zheng X, Chen Y, Li Z, Yang L. Identification of Renal Ischemia-Reperfusion Injury Subtypes and Predictive Model for Graft Loss after Kidney Transplantation Based on Programmed Cell Death-Related Genes. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:450-467. [PMID: 39664334 PMCID: PMC11631021 DOI: 10.1159/000540158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/26/2024] [Indexed: 12/13/2024]
Abstract
Introduction Ischemia-reperfusion injury (IRI) is detrimental to kidney transplants and may contribute to poor long-term outcomes of transplantation. Programmed cell death (PCD), a regulated cell death form triggered by IRI, is often indicative of an unfavorable prognosis following transplantation. However, given the intricate pathophysiology of IRI and the considerable variability in clinical conditions during kidney transplantation, the specific patterns of cell death within renal tissues remain ambiguous. Consequently, accurately predicting the outcomes for transplanted kidneys continues to be a formidable challenge. Methods Eight Gene Expression Omnibus datasets of biopsied transplanted kidney samples post-IRI and 1,548 PCD-related genes derived from 18 PCD patterns were collected in our study. Consensus clustering was performed to identify distinct IRI subtypes based on PCD features (IRI PCD subtypes). Differential enrichment analysis of cell death, metabolic signatures, and immune infiltration across these subtypes was evaluated. Three machine learning algorithms were used to identify PCD patterns related to prognosis. Genes associated with graft loss were screened for each PCD type. A predictive model for graft loss was constructed using 101 combinations of 10 machine learning algorithms. Results Four IRI subtypes were identified: PCD-A, PCD-B, PCD-C, and PCD-D. PCD-A, characterized by high enrichment of multiple cell death patterns, significant metabolic paralysis, and immune infiltration, showed the poorest prognosis among the four subtypes. While PCD-D involved the least kind of cell death patterns with the features of extensive activation of metabolic pathways and the lowest immune infiltration, correlating with the best prognosis in the four subtypes. Using various machine learning algorithms, 10 cell death patterns and 42 PCD-related genes were identified as positively correlated with graft loss. The predictive model demonstrated high sensitivity and specificity, with area under the curve values for 0.5-, 1-, 2-, 3-, and 4-year graft survival at 0.888, 0.91, 0.926, 0.923, and 0.923, respectively. Conclusion Our study explored the comprehensive features of PCD patterns in transplanted kidney samples post-IRI. The prediction model shows great promise in forecasting graft loss and could aid in risk stratification in patients following kidney transplantation.
Collapse
Affiliation(s)
- Jing Ji
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuan Ma
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xintong Liu
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Qingqing Zhou
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xizi Zheng
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Chen
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zehua Li
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Yang
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
45
|
He R, Liu Y, Fu W, He X, Liu S, Xiao D, Tao Y. Mechanisms and cross-talk of regulated cell death and their epigenetic modifications in tumor progression. Mol Cancer 2024; 23:267. [PMID: 39614268 PMCID: PMC11606237 DOI: 10.1186/s12943-024-02172-y] [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: 08/21/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024] Open
Abstract
Cell death is a fundamental part of life for metazoans. To maintain the balance between cell proliferation and metabolism of human bodies, a certain number of cells need to be removed regularly. Hence, the mechanisms of cell death have been preserved during the evolution of multicellular organisms. Tumorigenesis is closely related with exceptional inhibition of cell death. Mutations or defects in cell death-related genes block the elimination of abnormal cells and enhance the resistance of malignant cells to chemotherapy. Therefore, the investigation of cell death mechanisms enables the development of drugs that directly induce tumor cell death. In the guidelines updated by the Cell Death Nomenclature Committee (NCCD) in 2018, cell death was classified into 12 types according to morphological, biochemical and functional classification, including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, PARP-1 parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence and mitotic catastrophe. The mechanistic relationships between epigenetic controls and cell death in cancer progression were previously unclear. In this review, we will summarize the mechanisms of cell death pathways and corresponding epigenetic regulations. Also, we will explore the extensive interactions between these pathways and discuss the mechanisms of cell death in epigenetics which bring benefits to tumor therapy.
Collapse
Affiliation(s)
- Ruimin He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Yifan Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Weijie Fu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Xuan He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China.
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Furong Laboratory, Xiangya School of Medicine, Central South University, Hunan, 410078, China.
| |
Collapse
|
46
|
Song D, Cen Y, Qian Z, Wu XS, Rivera K, Wee TL, Demerdash OE, Chang K, Pappin D, Vakoc CR, Tonks NK. PTPN23-dependent ESCRT machinery functions as a cell death checkpoint. Nat Commun 2024; 15:10364. [PMID: 39609437 PMCID: PMC11604704 DOI: 10.1038/s41467-024-54749-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/20/2024] [Indexed: 11/30/2024] Open
Abstract
Cell death plasticity is crucial for modulating tissue homeostasis and immune responses, but our understanding of the molecular components that regulate cell death pathways to determine cell fate remains limited. Here, a CRISPR screen of acute myeloid leukemia cells identifies protein tyrosine phosphatase non-receptor type 23 (PTPN23) as essential for survival. Loss of PTPN23 activates nuclear factor-kappa B, apoptotic, necroptotic, and pyroptotic pathways by causing the accumulation of death receptors and toll-like receptors (TLRs) in endosomes. These effects are recapitulated by depletion of PTPN23 co-dependent genes in the endosomal sorting complex required for transport (ESCRT) pathway. Through proximity-dependent biotin labeling, we show that NAK-associated protein 1 interacts with PTPN23 to facilitate endosomal sorting of tumor necrosis factor receptor 1 (TNFR1), sensitizing cells to TNF-α-induced cytotoxicity. Our findings reveal PTPN23-dependent ESCRT machinery as a cell death checkpoint that regulates the spatiotemporal distribution of death receptors and TLRs to restrain multiple cell death pathways.
Collapse
MESH Headings
- Humans
- Endosomal Sorting Complexes Required for Transport/metabolism
- Endosomal Sorting Complexes Required for Transport/genetics
- Endosomes/metabolism
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Apoptosis
- NF-kappa B/metabolism
- Cell Death
- Protein Tyrosine Phosphatases, Non-Receptor/metabolism
- Protein Tyrosine Phosphatases, Non-Receptor/genetics
- Toll-Like Receptors/metabolism
- Tumor Necrosis Factor-alpha/metabolism
- Signal Transduction
- Cell Line, Tumor
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- HEK293 Cells
- Receptors, Death Domain/metabolism
Collapse
Affiliation(s)
- Dongyan Song
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Molecular and Cellular Biology Graduate Program, Stony Brook University, Stony Brook, NY, USA
| | - Yuxin Cen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Molecular and Cellular Biology Graduate Program, Stony Brook University, Stony Brook, NY, USA
| | - Zhe Qian
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Molecular and Cellular Biology Graduate Program, Stony Brook University, Stony Brook, NY, USA
| | - Xiaoli S Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, USA
| | - Keith Rivera
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Tse-Luen Wee
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Osama E Demerdash
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Kenneth Chang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Darryl Pappin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | | | - Nicholas K Tonks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA.
| |
Collapse
|
47
|
Wang S, Zhang Y. Construction of an immunogenic cell death-related LncRNA signature to predict the prognosis of patients with lung adenocarcinoma. BMC Med Genomics 2024; 17:277. [PMID: 39604972 PMCID: PMC11600735 DOI: 10.1186/s12920-024-02042-y] [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: 07/27/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is one of the most common malignant diseases worldwide. This study aimed to construct an immunogenic cell death (ICD)-related long non-coding RNA (lncRNA) signature to effectively predict the prognosis of LUAD. METHODS The RNA-sequencing and clinical data of LUAD were downloaded from The Cancer Genome Atlas (TCGA). Least absolute shrinkage and selection operator (LASSO) and stepwise multivariate Cox proportional hazard regression analysis were utilized to construct lncRNA signature. Then, the reliability of the signature was evaluated in the training, validation and whole cohorts. The differences in the immune landscape and drug sensitivity between the low- and high-risk groups were analyzed. Finally, the expression level of the selected ICD-related lncRNAs in LUAD cell lines via reverse transcription quantitative PCR (RT-qPCR). CCK-8 and transwell assays were performed to study biological function of AC245014.3. RESULTS A signature consisting of 5 ICD-related lncRNAs was constructed. Kaplan Meier (K-M) survival analysis showed shorter overall survival (OS) in high-risk group. The receiver operating characteristic (ROC) curves and Multivariate Cox regression analysis showed the signature was good predictive and independent prognostic factor in LUAD. Moreover, the high-risk group had a lower level of antitumor immunity and was less sensitive to some chemotherapeutics and targeted drugs. Finally, the expression level of selected ICD-related lncRNAs was validated in LUAD cell lines by RT-qPCR. Knockdown of AC245014.3 significantly suppressed LUAD proliferation, migration and invasion. CONCLUSIONS In this study, an ICD-related lncRNA signature was constructed, which could accurately predict the prognosis of LUAD patients and guide clinical treatment.
Collapse
Affiliation(s)
- Shuaishuai Wang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Yi Zhang
- Department of Orthopedic, Jinan Third People's Hospital, Jinan, Shandong, China
| |
Collapse
|
48
|
Yang Y, Zheng P, Duan B, Yang Y, Zheng X, Li W, Liu Q, Hu Y, Ma Y. A personalized vaccine combining immunogenic cell death-induced cells and nanosized antigens for enhanced antitumor immunity. J Control Release 2024; 376:1271-1287. [PMID: 39515613 DOI: 10.1016/j.jconrel.2024.10.060] [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: 07/04/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
The tumor vaccine aims to activate the immune system, promote antitumor cellular responses, and restore immune recognition and clearance of tumor cells. However, the low immunogenicity and heterogeneity of tumor antigens, along with immunosuppressive mechanisms, severely hinder tumor vaccines from achieving an efficient and sustained antitumor effect. Herein, we developed a combined vaccine strategy that utilizes immunogenic cell death (ICD) to elicit a broad spectrum of antigen-specific responses in a whole-cell-based manner. Additionally, we introduced nanosized antigens to intensify immune responses targeting a key tumor antigen. The combination of mitoxantrone (MTX) and curcumin (Cur) optimized ICD properties in TC-1 tumor cells, as evidenced by increased release of "find me" signals, such as HMGB1 and ATP, and enhanced exposure of the "eat me" signal, CALR, compared to either MTX or Cur alone. Correspondingly, the ICD cells induced by the combination produced more significant antitumor effects in vivo. Furthermore, the ICD cells in combination with E7-HBcAg VLPs or E7-Q11 nanofibers induced more intense effector cell responses to the antigen included in the nanovaccines, as well as a broad spectrum of antigens provided by tumor cells, and significantly suppressed the growth of established tumors compared with either ICD cells, VLPs, or nanofibers alone. In conclusion, the combination of ICD cells and nanosized antigens produced synergistic antitumor effects and elicited robust and comprehensive antitumor immunity, presenting an attractive strategy for developing personalized tumor vaccines.
Collapse
Affiliation(s)
- Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China; Cell Biology & Molecular Biology Laboratory of Experimental Teaching Center, Faculty of Basic Medical Science, Kunming Medical University, Chunrong West Road, Kunming 650500, China
| | - Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China; Kunming Medical University Graduate School, Chunrong West Road, Kunming 650500, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China
| | - Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China; Kunming Medical University Graduate School, Chunrong West Road, Kunming 650500, China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China; School of Life Sciences, Yunnan University, No.2 Cuihu North Road, Kunming 650091, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China; State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
49
|
Sargsian A, Koutsoumpou X, Girmatsion H, Egil C, Buttiens K, Luci CR, Soenen SJ, Manshian BB. Silver nanoparticle induced immunogenic cell death can improve immunotherapy. J Nanobiotechnology 2024; 22:691. [PMID: 39523339 PMCID: PMC11552147 DOI: 10.1186/s12951-024-02951-1] [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: 05/21/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Cancer immunotherapy is often hindered by an immunosuppressive tumor microenvironment (TME). Various strategies are being evaluated to shift the TME from an immunologically 'cold' to 'hot' tumor and hereby improve current immune checkpoint blockades (ICB). One particular hot topic is the use of combination therapies. Here, we set out to screen a variety of metallic nanoparticles and explored their in vitro toxicity against a series of tumor and non-tumor cell lines. For silver nanoparticles, we also explored the effects of core size and surface chemistry on cytotoxicity. Ag-citrate-5 nm nanoparticles were found to induce high cytotoxicity in Renca cells through excessive generation of reactive oxygen species (ROS) and significantly increased cytokine production. The induced toxicity resulted in a shift of the immunogenic cell death (ICD) marker calreticulin to the cell surface in vitro and in vivo. Subcutaneous Renca tumors were treated with anti-PD1 or in combination with Ag-citrate-5 nm. The combination group resulted in significant reduction in tumor size, increased necrosis, and immune cell infiltration at the tumor site. Inhibition of cytotoxic CD8 + T cells confirmed the involvement of these cells in the observed therapeutic effects. Our results suggest that Ag-citrate-5 nm is able to promote immune cell influx and increase tumor responsiveness to ICB therapies.
Collapse
Affiliation(s)
- Ara Sargsian
- NanoHealth and Optical Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Xanthippi Koutsoumpou
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Hermon Girmatsion
- NanoHealth and Optical Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Can Egil
- NanoHealth and Optical Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Kiana Buttiens
- NanoHealth and Optical Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Carla Rios Luci
- NanoHealth and Optical Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Stefaan J Soenen
- NanoHealth and Optical Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Bella B Manshian
- NanoHealth and Optical Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.
- Leuven Cancer Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
50
|
Xue D, Xu M, Madden MD, Lian X, Older EA, Pulliam C, Hui Y, Shang Z, Gupta G, Raja MK, Wang Y, Sardi A, Long Y, Chen H, Fan D, Bugni TS, Testerman TL, Wu Q, Li J. Discovery of A Chimeric Polyketide Family as Cancer Immunogenic Chemotherapeutic Leads. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.05.622009. [PMID: 39574732 PMCID: PMC11580922 DOI: 10.1101/2024.11.05.622009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Discovery of cancer immunogenic chemotherapeutics represents an emerging, highly promising direction for cancer treatment that uses a chemical drug to achieve the efficacy of both chemotherapy and immunotherapy. Herein we report a high-throughput screening platform and the subsequent discovery of a new class of cancer immunogenic chemotherapeutic leads. Our platform integrates informatics-based activity metabolomics for rapid identification of microbial natural products with both novel structures and potent activities. Additionally, we demonstrate the use of microcrystal electron diffraction (MicroED) for direct structure elucidation of the lead compounds from partially purified mixtures. Using this strategy to screen geographically and phylogenetically diverse microbial metabolites against pseudomyxoma peritonei, a rare and severe cancer, we discovered a new class of leads, aspercyclicins. The aspercyclicins feature an unprecedented tightly packed polycyclic polyketide scaffold that comprises continuous fused, bridged, and spiro rings. The biogenesis of aspercyclicins involves two distinct biosynthetic pathways, leading to formation of chimeric compounds that cannot be predicted by bottom-up approaches mining natural products biosynthetic genes. With comparable potency to some clinically used anticancer drugs, aspercyclicins are active against multiple cancer cell types by inducing immunogenic cell death (ICD), including the release of damage-associated molecular patterns and subsequent phagocytosis of cancer cells. The broad-spectrum ICD-inducing activity of aspercyclicins, combined with their low toxicity to normal cells, represents a new class of potential cancer immunogenic chemotherapeutics and particularly the first drug lead for pseudomyxoma peritonei treatment.
Collapse
|