1
|
Chen Z, Tai Y, Deng C, Sun Y, Chen H, Luo T, Lin J, Chen W, Xu H, Song G, Tang Q, Lu J, Zhu X, Wen S, Wang J. Innovative sarcoma therapy using multifaceted nano-PROTAC-induced EZH2 degradation and immunity enhancement. Biomaterials 2025; 321:123344. [PMID: 40262462 DOI: 10.1016/j.biomaterials.2025.123344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 04/05/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Sarcomas are highly malignant tumors characterized by their heterogeneity and resistance to conventional therapies, which significantly limit treatment options. EZH2 is highly expressed in sarcomas, but targeting it is difficult. In this study, we uncovered the non-canonical transcriptional mechanisms of EZH2 in sarcoma and highlighted the essential role of EZH2 in regulating YAP1 through non-canonical transcriptional pathways in the progression of sarcoma. Building on this, we developed YM@VBM, a novel and versatile nano-PROTAC (proteolysis-targeting chimera), by integrating a polyphenol-vanadium oxide system with the EZH2 degrader YM281 PROTAC, encapsulated in methoxy polyethylene glycol-NH2 to enhance biocompatibility. To further facilitate targeted drug delivery to tumors, YM@VBM nano-PROTACs were incorporated into microneedle patches. Our engineered YM@VBM exhibited multiple functionalities, including the peroxidase-like activity to generate reactive oxygen species, depletion of glutathione, and photothermal effects, specifically targeting sarcoma characteristics. YM@VBM significantly enhanced targeting efficacy via inducing potent EZH2 degradation. Most importantly, it can also activate anti-tumor immunity via excluding myeloid-derived suppressor cells, maturing dendritic cells, and forming tertiary lymphoid structures. Hence, we reveal that YM@VBM presents a promising treatment strategy for sarcoma, offering a multifaceted approach to combat this challenging malignancy.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Yi Tai
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China; Surgical Department of Colorectal Cancer, Zhejiang Cancer Hospital, 1st BanShan East Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, PR China
| | - Chuangzhong Deng
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Yameng Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Hongmin Chen
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Tianqi Luo
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Jiaming Lin
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Weiqing Chen
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Huaiyuan Xu
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Guohui Song
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Qinglian Tang
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Jinchang Lu
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Xiaojun Zhu
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China.
| | - Jin Wang
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China.
| |
Collapse
|
2
|
Li J, Zhang G, Sun Z, Jiang M, Jia G, Liu H, Liu N, Shi L, Zhang L, Nie L, Zhang Y, Fu Y. Immunogenic cuproptosis in cancer immunotherapy via an in situ cuproptosis-inducing system. Biomaterials 2025; 319:123201. [PMID: 40020502 DOI: 10.1016/j.biomaterials.2025.123201] [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: 12/14/2024] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 03/03/2025]
Abstract
Cell death-based therapies combined with immunotherapy have great potential in cancer therapy. To further explore and apply the combined therapies, the immunogenicity of different cell death modes in colorectal cancer (CRC) was evaluated by a cause-and-effect framework encompassing 12 cell death modes. Results show robust correlations among cuproptosis, immunogenic cell death (ICD) and immunity in CRC, as observed in our in-house and other independent cohorts, which are substantiated by in vitro and in vivo experiments. Subsequent investigations demonstrate that cuproptosis induces endoplasmic reticulum stress, leading to the release of damage-associated molecular patterns from CRC cells and triggering the maturation of antigen-presenting cells. Moreover, for in vivo therapeutic approaches, an in situ cuproptosis-inducing system was devised, which can further strengthen the effects of immune cells. Through the combined analysis including single-cell RNA sequencing, cuproptosis is shown to mobilize cytotoxic T lymphocytes and M1 macrophages within the tumor microenvironment (TME). Additionally, co-treatment with Imiquimod, the TLR7 agonist, augments the anti-tumor immune responses induced by cuproptosis. Overall, we provide compelling evidence that cuproptosis induces ICD thus fostering an inflammatory TME, and the cuproptosis-based delivery system further promotes this inflammatory environment, demonstrating considerable potential for enhancing tumor therapy efficacy.
Collapse
Affiliation(s)
- Jiehan Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Ge Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Zhao Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Meimei Jiang
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Guiyun Jia
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Hao Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Nannan Liu
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Liyang Shi
- College of Biology, Hunan University, Changsha, 410082, China
| | - Lingling Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Liming Nie
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Yingjie Zhang
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China.
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, 450052, China.
| |
Collapse
|
3
|
Tan K, Zhang H, Yang J, Wang H, Li Y, Ding G, Gu P, Yang S, Li J, Fan X. Organelle-oriented nanomedicines in tumor therapy: Targeting, escaping, or collaborating? Bioact Mater 2025; 49:291-339. [PMID: 40161442 PMCID: PMC11953998 DOI: 10.1016/j.bioactmat.2025.02.040] [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: 12/25/2024] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Precise tumor therapy is essential for improving treatment specificity, enhancing efficacy, and minimizing side effects. Targeting organelles is a key strategy for achieving this goal and is a frontier research area attracting a considerable amount of attention. The concept of organelle targeting has a significant effect on the structural design of the nanodrugs employed. Most notably, the intricate interactions among different organelles in a tumor cell essentially create a unified system. Unfortunately, this aspect might have been somewhat overlooked when existing organelle-targeting nanodrugs were designed. In this review, we underscore the synergistic relationship among the various organelles and advocate for a holistic view of organelle-targeting design. Through the integration of biology and material science, recent advancements in organelle targeting, escaping, and collaborating are consolidated to offer fresh perspectives for the development of antitumor nanomedicines.
Collapse
Affiliation(s)
- Kexin Tan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Haiyang Zhang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Jianyuan Yang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Hang Wang
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Yongqiang Li
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Guqiao Ding
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Ping Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Siwei Yang
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Jipeng Li
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| |
Collapse
|
4
|
Gabizon AA. Cancer nanomedicine from a clinician-scientist perspective: Lessons and prospects. J Control Release 2025; 382:113731. [PMID: 40228664 DOI: 10.1016/j.jconrel.2025.113731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
The nanomedicine field has progressed enormously in the last couple of decades. From a loose group of liposomologists, polymer scientists, chemical engineers, and experts in metal nanoparticles, mesoporous silica, and other nanomaterials, the field has gradually consolidated and has generated vast amounts of research and clinical data, but, until the development of lipid nanoparticle (LNP)-based vaccinations for Covid-19, has remained with low visibility in the clinic. Applications in the cancer field are the most frequently sought projects in nanomedicine. For the last 45 years, my clinical career has mingled with my research career focusing on ways to formulate drugs in liposomes to improve their safety and efficacy in cancer therapy. In this review, I will discuss my contribution to the development of pegylated liposomal doxorubicin and other cancer nanomedicines from my privileged position as a clinician and scientist.
Collapse
Affiliation(s)
- Alberto A Gabizon
- The Leah and Jakub Susskind Nano-Oncology Research Laboratory at the Helmsley Cancer Center, Shaare Zedek Medical Center and the Hebrew University-Faculty of Medicine, Jerusalem, Israel.
| |
Collapse
|
5
|
Li Y, Chen Y, Tang Y, Yang T, Zhou P, Miao L, Chen H, Deng Y. Breaking the barriers in effective and safe Toll-like receptor stimulation via nano-immunomodulators for potent cancer immunotherapy. J Control Release 2025; 382:113667. [PMID: 40157608 DOI: 10.1016/j.jconrel.2025.113667] [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: 12/02/2024] [Revised: 02/20/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Immunotherapy is an emerging strategy that awakens the intrinsic immune system for cancer treatment. Generally, successful immunotherapy of malignant tumours relies on the effective production of tumour-associated antigens and their lymph node delivery, antigen processing and presentation for T-cell activation, and the dismantling of the immunosuppressive tumour microenvironment. Toll-like receptor (TLR) agonists are potent stimulants in cancer immunotherapy, which can directly activate antigen-presenting cells (APCs) and further induce T cell activation for antitumour immune response and convert immunosuppressive tumour microenvironment to an immunogenic one for cooperative tumour ablation. However, TLR agonists for effective cancer immunotherapy have encountered essential challenges, such as insufficient immune activation and systemic side effects. In recent years, nano-immunomodulators with TLR agonists have been employed for tumour- and/or lymph node-targeted immune activation to improve the antitumour immune response and alleviate their systemic toxicities, providing a promising strategy for enhanced cancer immunotherapy. Herein, we introduce the recent progress in developing various TLR nano-immunomodulators for cancer immunotherapy via APC activation and tumour microenvironment remodelling. Upon elucidating the rational design principles of nano-immunomodulators, we elucidate the advancement of TLR nanoagonists to break the barriers in effective and safe Toll-like receptor stimulation for potent cancer immunotherapy.
Collapse
Affiliation(s)
- Yaoqi Li
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yitian Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yong'an Tang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Tao Yang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ping Zhou
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou 215006, China.
| | - Huabing Chen
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China.
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China; State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China.
| |
Collapse
|
6
|
Hou X, Wang C, Zhong Y, Wang L, Kang DD, Lubitz G, Xue Y, Liu Z, Wang S, Li H, Tian M, Cao D, Guo K, Deng B, McComb DW, Marron TU, Brown BD, Merad M, Brody JD, Dong Y. Enhancing antitumor immunity through chemotherapeutic-derived lipid nanoparticle-induced immunogenic cell death and CD40L/Flt3L mRNA-mediated dendritic cell activation. J Control Release 2025; 382:113684. [PMID: 40185331 DOI: 10.1016/j.jconrel.2025.113684] [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: 12/30/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Dendritic cells (DCs) are essential for inducing effective antitumor T cell responses. However, the immunosuppressive tumor microenvironment (TME) hinders DC recruitment and maturation, facilitating tumor progression and spread. This study investigates the synergistic potential of immunogenic cell death (ICD), triggered by chemotherapeutic-derived lipid nanoparticles (LNPs), in combination with Flt3L and CD40L mRNA delivery to enhance DC mobilization and activation, reprogram the TME, and ultimately promote robust antitumor T cell responses. The optimized LNP formulation, GEM5Q7, efficiently delivered mRNA and induced ICD in melanoma cells. Intratumoral administration of GEM5Q7, encapsulating Flt3L and CD40L mRNAs, elevated pro-inflammatory cytokine and chemokine secretion, driving the infiltration and activation of cross-presenting DCs, which are critical for priming T cells. In a subcutaneous melanoma model, this approach led to significant tumor suppression and a 40 % complete response rate. This strategy holds promise for enhancing cancer immunotherapies by reprogramming the TME and inducing durable antitumor T cell immunity.
Collapse
Affiliation(s)
- Xucheng Hou
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chang Wang
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yichen Zhong
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Leiming Wang
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Diana D Kang
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Gabrielle Lubitz
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yonger Xue
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhengwei Liu
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Siyu Wang
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Haoyuan Li
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Meng Tian
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dinglingge Cao
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kaiyuan Guo
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Binbin Deng
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH 43212, USA
| | - David W McComb
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH 43212, USA
| | - Thomas U Marron
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian D Brown
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joshua D Brody
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Yizhou Dong
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
7
|
Zhang H, Xu X, Li S, Huang H, Zhang K, Li W, Wang X, Yang J, Yin X, Qu C, Ni J, Dong X. Advances in nanoplatform-based multimodal combination therapy activating STING pathway for enhanced anti-tumor immunotherapy. Colloids Surf B Biointerfaces 2025; 250:114573. [PMID: 39983453 DOI: 10.1016/j.colsurfb.2025.114573] [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: 09/25/2024] [Revised: 01/24/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
Activation of the cyclic GMP-AMP synthase(cGAS)-stimulator of interferon genes (STING) has great potential to promote antitumor immunity. As a major effector of the cell to sense and respond to the aberrant presence of cytoplasmic double-stranded DNA (dsDNA), inducing the expression and secretion of type I interferons (IFN) and STING, cGAS-STING signaling pathway establishes an effective natural immune response, which is one of the fundamental mechanisms of host defense in organisms. In addition to the release of heterologous DNA due to pathogen invasion and replication, mitochondrial damage and massive cell death can also cause abnormal leakage of the body's own dsDNA, which is then recognized by the DNA receptor cGAS and activates the cGAS-STING signaling pathway. However, small molecule STING agonists suffer from rapid excretion, low bioavailability, non-specificity and adverse effects, which limits their therapeutic efficacy and in vivo application. Various types of nano-delivery systems, on the other hand, make use of the different unique structures and surface modifications of nanoparticles to circumvent the defects of small molecule STING agonists such as fast metabolism and low bioavailability. Also, the nanoparticles are precisely directed to the focal site, with their own appropriate particle size combined with the characteristics of passive or active targeting. Herein, combined with the cGAS-STING pathway to activate the immune system and kill tumor tissues directly or indirectly, which help maximize the use of the functions of chemotherapy, photothermal therapy(PTT), chemodynamic therapy(CDT), and radiotherapy(RT). In this review, we will discuss the mechanism of action of the cGAS-STING pathway and introduce nanoparticle-mediated tumor combination therapy based on the STING pathway. Collectively, the effective multimodal nanoplatform, which can activate cGAS-STING pathway for enhanced anti-tumor immunotherapy, has promising avenue clinical applications for cancer treatment.
Collapse
Affiliation(s)
- Huizhong Zhang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaohan Xu
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shiman Li
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huating Huang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ke Zhang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wenjing Li
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xinzhu Wang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingwen Yang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xingbin Yin
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Changhai Qu
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Ni
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xiaoxv Dong
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
8
|
Wang B, Tang X, Xiao C, Yu Z, Bo H, Wang J, Wang J. Nucleus-targeted ruthenium(II) complex triggers immunogenic cell death and sensitizes melanoma to anti-PD-1 therapy by activating cGAS-STING pathway. J Inorg Biochem 2025; 267:112871. [PMID: 40022761 DOI: 10.1016/j.jinorgbio.2025.112871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
A significant challenge in the treatment of melanoma with immune checkpoint blockades (ICBs) is the limited T cells response often observed in immunologically "cold" tumors. By leveraging the immunogenicity of immunogenic cell death (ICD), which increases the susceptibility of tumor cells to ICBs, this study investigated the potential of a nucleus-targeted ruthenium(II) complex (Ru1) as an inducer of ICD. Treatment with Ru1 induced DNA damage in melanoma cells, activating the cyclic GMP-AMP synthase-stimulator of the interferon genes (cGAS-STING) pathway. This triggered endoplasmic reticulum (ER) stress, leading to ICD. Ru1-treated dying melanoma cells exhibited characteristics such as cell exposure of calreticulin (CRT) on the cell surface, release of adenosine triphosphate (ATP), and secretion of high-mobility group box 1 (HMGB1). Vaccination with Ru1-treated, dying melanoma cells elicited robust antitumor immune responses, as evidenced by CD8+ T cells activation, reduced Foxp3+ T cells count, and the development of a memory immune response that protected mice from subsequent melanoma challenges. Combining Ru1 with anti-PD-1 therapy significantly promoted T cells infiltration, enhanced dendritic cell activation, and reduced tumor-associated immunosuppressive factors, indicating a reprogramming of the tumor microenvironment. These findings suggest that Ru1 is a promising therapeutic agent for treating "cold" tumors in cancer chemoimmunotherapy.
Collapse
Affiliation(s)
- Bishu Wang
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Xingguo Tang
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Chuntao Xiao
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Zhijie Yu
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Huaben Bo
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Jie Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Jinquan Wang
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| |
Collapse
|
9
|
Chen R, Ren J, Wang Y, Zhang X, Jia Y, Liu C, Qu K. A Comprehensive Analysis Exploring the Impact of an Immunogenic Cell Death-Related Panel for Ovarian Cancer. Mol Biotechnol 2025; 67:2520-2535. [PMID: 39112745 PMCID: PMC12055628 DOI: 10.1007/s12033-024-01215-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/02/2024] [Indexed: 05/07/2025]
Abstract
Ovarian cancer (OV) is a malignant tumor that ranks first among gynecological cancers, thus posing a significant threat to women's health. Immunogenic cell death (ICD) can regulate cell death by activating the adaptive immune system. Here, we aimed to comprehensively characterize the features of ICD-associated genes in ovarian cancer, and to investigate their prognostic value and role in the response to immunotherapy. After analyzing datasets from The Cancer Genome Atlas, we utilized weighted gene coexpression network analysis to screen for hub genes strongly correlated with ICD genes in OV, which was subsequently validated with OV samples from the Gene Expression Omnibus (GEO) database. A prognostic risk model was then constructed after combining univariate, multivariate Cox regression and LASSO regression analysis to recognize nine ICD-associated molecules. Next, we stratified all OV patients into two subgroups according to the median value. The multivariate Cox regression analysis showed that the risk model could predict OV patient survival with good accuracy. The same results were also found in the validation set from GEO. We then compared the degree of immune cell infiltration in the tumor microenvironment between the two subgroups of OV patients, and revealed that the high-risk subtype had a higher degree of immune infiltration than the low-risk subtype. Additionally, in contrast to patients in the high-risk subgroup, those in the low-risk subgroup were more susceptible to chemotherapy. In conclusion, our research offers an independent and validated model concerning ICD-related molecules to estimate the prognosis, degree of immune infiltration, and chemotherapy susceptibility in patients with OV.
Collapse
Affiliation(s)
- Rui Chen
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, China
| | - Jie Ren
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, China
| | - Yifei Wang
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, China
| | - Xing Zhang
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, China
- Hangzhou Institute of National Extremely-Weak Magnetic Field Infrastructure, Hangzhou, 310028, China
| | - Yifan Jia
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, China.
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Chang Liu
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, China.
| | - Kai Qu
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, China.
| |
Collapse
|
10
|
Mannis GN, Abboud CN, Daver NG, Murthy GSG, Wang ES, Bradley TJ, Yaghmour G, Vachhani P, Balasubramanian SK, Chua CC, Fong CY, Asch AS, Dong M, Li S, Bagheri T, Doshi P, Vyas P, Malki MMA. Phase 2 Multi-Arm Study of Magrolimab Combinations in Patients With Acute Myeloid Leukaemia. EJHAEM 2025; 6:e70051. [PMID: 40364806 PMCID: PMC12073901 DOI: 10.1002/jha2.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Accepted: 04/13/2025] [Indexed: 05/15/2025]
Abstract
Introduction This phase 2 study evaluated magrolimab+venetoclax (VEN)+azacitidine (AZA) in untreated, unfit acute myeloid leukaemia (AML) and magrolimab+mitoxantrone+etoposide+cytarabine in relapsed/refractory (R/R) AML. Methods Endpoints included complete remission rate (CRR), overall response rate (ORR), overall survival (OS) and safety. Results Eighteen and 36 patients were enrolled into the unfit and R/R AML arms, respectively. CRR was 38.9% and 25.0%, ORR was 66.7% and 38.9%, and median OS was 15.3 and 10.5 months in the unfit AML and R/R AML arms, respectively. No dose-limiting toxicities or magrolimab-related deaths occurred. Conclusion Magrolimab was safely combined with existing AML therapies with no new safety signals. Clinical Trial Registration This trail was registered at www.clinicaltrials.gov as NCT04778410.
Collapse
Affiliation(s)
| | | | - Naval G. Daver
- The University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | | | - Eunice S. Wang
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Terrence J. Bradley
- Sylvester Comprehensive Cancer CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - George Yaghmour
- University of Southern California Norris Comprehensive Cancer CenterLos AngelesCaliforniaUSA
| | - Pankit Vachhani
- O'Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | | | | | - Chun Yew Fong
- Austin Hospital and University of MelbourneMelbourneVictoriaAustralia
| | - Adam S. Asch
- Stephenson Cancer CenterUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Mei Dong
- Gilead Sciences, Inc.Foster CityCaliforniaUSA
| | - Shuang Li
- Gilead Sciences, Inc.Foster CityCaliforniaUSA
| | | | - Parul Doshi
- Gilead Sciences, Inc.Foster CityCaliforniaUSA
| | - Paresh Vyas
- Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford and Department of HaematologyOxford University Hospitals NHS TrustOxfordUK
| | | |
Collapse
|
11
|
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 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] [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
|
12
|
Engelen Y, Demuynck R, Ramon J, Breckpot K, De Smedt S, Lajoinie GPR, Braeckmans K, Krysko DV, Lentacker I. Immunogenic cell death as interplay between physical anticancer modalities and immunotherapy. J Control Release 2025:113721. [PMID: 40368187 DOI: 10.1016/j.jconrel.2025.113721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
Current cancer treatment strategies in practice nowadays often face limitations in effectiveness due to factors such as resistance, recurrence, or suboptimal outcomes. Traditional approaches like chemotherapy often come with severe systemic side effects due to their non-specific action, prompting the development of more targeted therapies. Among these, physical ablation techniques such as radiotherapy (RT) and focused ultrasound (FUS) have gained attention for their ability to precisely target malignant tissues, reduce physical and mental stress for the patients, and minimize recovery time. These therapies also aim to stimulate the immune system through a process referred to as immunogenic cell death (ICD), enhancing the body's ability to fight cancer, explaining abscopal effects. RT has been the most established of the abovementioned techniques for decades, and will not be included in the review. While initially focused on complete tumor ablation, these techniques are now shifting towards milder, more controlled applications that induce ICD without extensive tissue damage. This review explores how physical ablation therapies can harness ICD to boost anticancer immunity, emphasizing their potential to complement immunotherapies and improve outcomes for cancer patients.
Collapse
Affiliation(s)
- Y Engelen
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
| | - R Demuynck
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - J Ramon
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium
| | - K Breckpot
- Laboratory for Molecular and Cellular Therapy, Translational Oncology Research Center, Department of Biomedical Sciences, Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - S De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - G P R Lajoinie
- Physics of Fluids Group, Technical Medical (TechMed) Center, and Max Planck Center for Complex Fluid Dynamics, University of Twente, Enschede, the Netherlands
| | - K Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium
| | - D V Krysko
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - I Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
13
|
Luo K, You S, Chen J, Ye W, Tian J, Feng X, Wang Y, Li L, Yu X. Construction of multifunctional nanozymes with amplified immunogenic death effect as a long-term anti-tumor nanoplatform. BIOMATERIALS ADVANCES 2025; 175:214336. [PMID: 40344986 DOI: 10.1016/j.bioadv.2025.214336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/22/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Stimulating the immunogenic cell death (ICD) effect is a method of tumor treatment through activating immunity. While conventional approaches including chemotherapy, photothermal therapy (PTT), and chemodynamic therapy (CDT), demonstrate partial ICD induction capabilities, their efficacy in eliciting systemic immune responses remains constrained by the immunosuppressive tumor microenvironment. Herein, a trimetallic nanozyme (Mn/Fe-MIL-101/CuS/DOX@FA) was engineered through the integration of Mn-doped Fe-MOFs, CuS, doxorubicin (DOX, 35.08 mg/g), and folic acid (FA). The design leverages the synergistic effects of Mn(II), Fe(III), and Cu(II), combined with the photothermal performance of CuS, which collectively enhance glutathione peroxidase (GPx)-like and peroxidase (POD)-like activities. This catalytic cascade depletes glutathione and boosts hydroxyl radicals via Fenton-like reactions, thereby disrupting redox balance to amplify chemodynamic therapy. CuS-mediated photothermal effects coupled with pH/GSH-responsive DOX release further augment ICD, effectively reversing immunosuppression. In vivo evaluations demonstrated 57 % inhibition of the primary tumor and 66.7 % inhibition of the distant tumor, confirming its efficacy in tumor treatment and prevention of recurrence/metastasis. Besides, magnetic resonance imaging experiments showed the T1/T2 dual-mode imaging performance. Thereby, a long-term anti-tumor nanoplatform is constructed through dual-mode imaging-guided multimodal therapy, which integrates tumor diagnosis, treatment, and prevention of recurrence and metastasis.
Collapse
Affiliation(s)
- Kexin Luo
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei University, Wuhan 430062, China
| | - Sasha You
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei University, Wuhan 430062, China
| | - Jingyu Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio-resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Wang Ye
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei University, Wuhan 430062, China
| | - Jian Tian
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei University, Wuhan 430062, China
| | - Xiyue Feng
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei University, Wuhan 430062, China
| | - Yingxi Wang
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei University, Wuhan 430062, China
| | - Ling Li
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals & Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei University, Wuhan 430062, China.
| | - Xiaolan Yu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio-resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, China.
| |
Collapse
|
14
|
Gautam S, Joshi S, Jindal P, Patel P, Pal RR, Kumar M, Gupta GD, Kurmi BD. Recent doxorubicin-conjugates in cancer drug delivery: Exploring conjugation strategies for enhanced efficacy and reduced toxicity. Int J Pharm 2025; 675:125556. [PMID: 40187698 DOI: 10.1016/j.ijpharm.2025.125556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Doxorubicin is a first-line treatment of cancer that works on the mechanism of DNA intercalation and topoisomerase II poisoning. Since the 20th century, Doxorubicin has been used as a promising drug to treat several types of cancer, both solid or metastatic, including breast, thyroid, bladder, ovarian, or gastric cancer, etc. Even though it shows promising effects on cancer cells, it also shows its effects on healthy cells with cancerous cells, which leads to several severe side effects, such as cardiomyopathy, phlebitis, congestive heart failure (CHF), etc., which limits its usage in chemotherapy. Several research has focused on the targeted delivery of doxorubicin to cancerous cells to reduce side effects and improve efficacy. To optimize its anticancer potential, scientists have recently been developing nano-formulations and investigating various conjugations. The structure of doxorubicin consists of two primary functional groups that can be employed for conjugation with a variety of biomolecules, The first is the primary amine group in a sugar moiety, and the other one is the primary hydroxyl group in the aliphatic chain ring. In this paper, we have mentioned several conjugations of doxorubicin such as antibodies, nanoparticles, polymers, and phytochemical conjugations. Different studies regarding these conjugations are also mentioned, which represent promising strategies to optimize cancer treatment by minimizing side effects.
Collapse
Affiliation(s)
- Shreastha Gautam
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Sachin Joshi
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Priya Jindal
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road Moga-142001, Punjab, India.
| | - Ravi Raj Pal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India.
| |
Collapse
|
15
|
Zhong H, Liang J, Xu X, Ding C, Yu M, Abuduaini N, Liu J, Wang X, Zhang S, Wang F, Feng B. Hematoporphyrin-Modified Dendrimers Combined Immunoadjuvants for Enhanced Photoimmunotherapy of Colorectal Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:25059-25070. [PMID: 40257172 DOI: 10.1021/acsami.5c02413] [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: 04/22/2025]
Abstract
Photoimmunotherapy has emerged as a promising strategy for cancer therapy due to its increased therapeutic effect, ability to reverse drug resistance, and enhanced immune activation. But there is still a lack of effective nanomaterial-based photothermal therapy (PTT) or photodynamic therapy (PDT) agents in photoimmunotherapy. In this study, photosensitizer hematoporphyrin-modified G5 PAMAM (G5-HP) nanomaterials are synthesized, which exhibit excellent photothermal conversion capability and photodynamic effects under 660 nm irradiation, effectively inducing tumor cell ablation and immunogenic cell death (ICD). Besides, ICD induced by G5-HP can generate tumor-associated antigens, thereby enhancing dendritic cell (DC) maturation and subsequent T cell activation. In addition, G5-HP polymers can bind to Toll-like receptor (TLR) agonists CpG-ODN through electrostatic interaction, forming stable G5-HP/CpG nanoparticles. The incorporation of CpG-ODN as an immunoadjuvant further amplified DC maturation, synergizing with phototherapy to strengthen antitumor immunity. Notably, in vivo studies confirmed that G5-HP/CpG nanoparticles significantly suppressed colorectal tumor growth under laser irradiation, while maintaining excellent biocompatibility. Taken together, the synthesized G5-HP polymers perform excellent PTT and PDT efficacy, and the formed G5-HP/CpG nanoparticles effectively integrate phototherapy with DC-mediated immunotherapy. This study offers a promising strategy for colorectal cancer treatment, leveraging the synergistic effects of phototherapy and immunotherapy to achieve superior antitumor outcomes.
Collapse
Affiliation(s)
- Hao Zhong
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Liang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ximo Xu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Chengsheng Ding
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Mengqin Yu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Naijipu Abuduaini
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jingyi Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaohan Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sen Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fei Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bo Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
16
|
Lei S, Li J, Zhu M, Zhou W, Fu X, Wu S, Chen X, Zhang J, Duan X, Wang W, Men K. Chimeric Antigen Receptor-Engineered Cell Membrane-Coated Nanoparticles Promote Dual-Targeted mRNA-Based Cancer Gene Therapy. ACS NANO 2025; 19:15668-15684. [PMID: 40227438 DOI: 10.1021/acsnano.4c18153] [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: 04/15/2025]
Abstract
Gene therapy using mRNA has facilitated progress in cancer therapy. However, its application is hindered by a limited tumor-targeted delivery approach, leading to off-target effects and safety concerns. Chimeric antigen receptor (CAR) molecules enable T cells to recognize specific antigens in a major histocompatibility complex-unrestricted manner. CAR approaches provide an "off-the-shelf" solution for introducing additional targeting functionality to a cell membrane. Cancer cell membrane-coated nanoparticles with homotypic tumor-targeted properties provide a readily accessible platform for gene engineering and membrane extraction. Herein, we demonstrate a CAR-inspired cancer cell membrane-coated platform for delivering an mRNA formulation through a dual tumor-targeted mechanism. The simplified human epidermal growth factor receptor 2 (HER2)-specific CAR molecule (comprising an extracellular HER2-binding domain, a hinge, and a transmembrane domain) was engineered on the cell membrane of cancer cells to establish CAR-CT26 cells. The extracted CAR-CT26 membrane (CARM) was subsequently coated onto the lipid nanoparticle (LNP)-mRNA surface to form a CARM@LNP-mRNA complex. In vitro, the CARM-coated nanoparticles exhibited enhanced mRNA transfection efficiency toward CT26 cells overexpressing target HER2 antigens. Systemic administration of the CARM@LNP-mRNA formulation resulted in stronger tumor-targeting ability and tumor suppression in HER2+ CT26 subcutaneous tumors and peritoneal cavity metastasis models than that observed with the CT26 cell membrane-coated version. Our data suggest that CARM@LNP is a feasible choice for mRNA-based gene therapy. These results provide evidence for the systemic administration of CARM@LNP-mRNA as a promising tumor-targeted therapeutic strategy.
Collapse
Affiliation(s)
- Sibei Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Pharmacy, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Jingmei Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Manfang Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Weilin Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xizi Fu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shan Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiayu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Wei Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ke Men
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
17
|
Huang H, Tong QS, Zhang JY, Miao WM, Yu HH, Wang J, Shen S, Du JZ. Phagocytosis-Activating Nanocomplex Orchestrates Macrophage-Mediated Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2500982. [PMID: 40289887 DOI: 10.1002/adma.202500982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/09/2025] [Indexed: 04/30/2025]
Abstract
The phagocytosis of macrophages to tumor cells represents an alluring strategy for cancer immunotherapy; however, its effectiveness is largely hindered by the detrimental upregulation of anti-phagocytic signals and insufficient expression of pro-phagocytic signals of tumor cells. Here, a pro-phagocytic polymer-based nanocomplex is designed to promote the macrophage engulfment of tumor cells through concurrent modulation of both the "eat me" and "don't eat me" signals. The nanocomplex MNCCD47i-CALRt is formed by complexing a synthetic PAMAM derivative (G4P-C7A) that is capable of intrinsically inducing the exposure of calreticulin (CALR, a crucial pro-phagocytic protein) and a small inference RNA that can inhibit the expression of CD47 (a primary anti-phagocytic protein). MNCCD47i-CALRt can significantly delay tumor growth and prolong the survival of tumor-bearing mice with negligible hematopoietic toxicity in multiple murine colorectal cancer models. Furthermore, the pro-phagocytic capacity of MNCCD47i-CALRt is validated in the patient-derived tumor organoid model. Collectively, the phagocytosis-promoting nanocomplex provides a simple and potent strategy for boosting macrophage-mediated cancer immunotherapy.
Collapse
Affiliation(s)
- Hua Huang
- College & Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, 230032, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Qi-Song Tong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Jing-Yang Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Wei-Min Miao
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Hui-Han Yu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| |
Collapse
|
18
|
Guilbaud E, Naulin F, Meziani L, Deutsch E, Galluzzi L. Impact of radiation therapy on the immunological tumor microenvironment. Cell Chem Biol 2025:S2451-9456(25)00099-6. [PMID: 40280118 DOI: 10.1016/j.chembiol.2025.04.001] [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: 12/29/2024] [Revised: 02/22/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Abstract
External beam radiation therapy (RT) is a cornerstone of modern cancer management, being utilized in both curative and palliative settings due to its safety, efficacy, and widespread availability. A primary biological effect of RT is DNA damage, which leads to significant cytostatic and cytotoxic effects. Importantly, malignant cells possess a limited capacity for DNA repair compared to normal cells, and when combined with irradiation techniques that minimize damage to healthy tissues, this creates an advantageous therapeutic window. However, the clinical effectiveness of RT also appears to involve both direct and indirect interactions between RT and non-transformed components of the tumoral ecosystem, particularly immune cells. In this review, we describe the molecular and cellular mechanisms by which irradiated cancer cells modify the immunological tumor microenvironment and how such changes ultimately impact tumor growth.
Collapse
Affiliation(s)
- Emma Guilbaud
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Flavie Naulin
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France
| | - Lydia Meziani
- Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France
| | - Eric Deutsch
- Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France.
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Chen M, Chen Y, Chen W, Chen X, Guo X, Yu J, Guo X, Wang M, Zhang X, Hu Q, Fang S, Zheng L, Zhao Z, Du Y, Shu G, Ji J. Immune-activated microspheres for enhanced chemoembolization of hepatocellular carcinoma by blocking the adenosine A2A receptor. Acta Biomater 2025:S1742-7061(25)00295-8. [PMID: 40274057 DOI: 10.1016/j.actbio.2025.04.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Transcatheter arterial chemoembolization (TACE) stands as the frontline strategy for unresectable hepatocellular carcinoma (HCC), effectively eliminating cancer cells through direct cytotoxicity and immunogenic cell death (ICD). However, TACE triggers rapid tumor apoptosis, which promotes the release of intracellular ATP into the extracellular space. This ATP is sequentially hydrolyzed to adenosine (ADO) by ectonucleotidases (CD39 and CD73) overexpressed in the tumor microenvironment (TME), resulting in ADO accumulation. The ensuing ADO pathway-mediated immunosuppression via adenosine 2A receptors (A2AR) signaling severely limits TACE-induced ICD efficacy, resulting in poor prognosis. To address this, we developed gelatin microspheres co-loaded with doxorubicin (DOX) and the A2AR antagonist SCH-58,261, in which SCH-58,261 was loaded into solid lipid nanoparticle (SLNP) due to its poor water solubility. The microspheres (SLNP-SCH/DOX@MS) showed an average size of 49 ± 13 μm, with the capable of complete tumor vascular embolization, and sustained release profiles of both DOX and SCH-58,261 over 30 days. In vitro and in vivo studies indicated that SLNP-SCH/DOX@MS not only enhanced tumor cell apoptosis but also amplified ICD-mediated dendritic cell maturation and antigen presentation. Moreover, SCH-58,261 counteracted TACE-triggered ADO accumulation by competitively binding to A2AR on immune cells, thereby reversing dendritic cell dysfunction and CD8+T cell exhaustion. This dual-action strategy synergized ICD-driven immunostimulation with ADO pathway blockade, reshaping the TME. Our findings highlight the potential of SLNP-SCH/DOX@MS to address the delicate equilibrium between ICD-induced immunity and ADO-mediated immunosuppression for improved HCC treatment. STATEMENT OF SIGNIFICANCE: This study introduces a approach to improve transcatheter arterial chemoembolization (TACE) for unresectable hepatocellular carcinoma (HCC) by addressing the adenosine (ADO) pathway, a known barrier to effective immunogenic cell death (ICD). We developed gelatin microspheres co-loaded with doxorubicin (DOX) and the A2AR antagonist SCH-58,261, which significantly enhance TACE-induced immunity by promoting ICD and counteracting ADO-mediated immunosuppression. In vitro and in vivo results demonstrate robust dendritic cell maturation and amplified tumor-specific immune responses, indicating improved antitumor efficacy. This work provides a promising strategy to optimize TACE for HCC treatment, offering our readership a therapeutic solution that bridges cancer treatment and immunomodulation.
Collapse
Affiliation(s)
- Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China; Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Yaning Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China
| | - Weiqian Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China; Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiaoxiao Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China; Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiaoju Guo
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China; Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Junchao Yu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China; Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xinyu Guo
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China; Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Mengyuan Wang
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Xinyu Zhang
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Qin Hu
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Shiji Fang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China; Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Liyun Zheng
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China; Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China; Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, China.
| | - Gaofeng Shu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China; Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| | - Jiansong Ji
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, School of Medicine, Lishui Hospital of Zhejiang University, Zhejiang, China; Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China; School of Medicine, Lishui University, Zhejiang, China.
| |
Collapse
|
20
|
Tang Y, Xiang D, Li Q. In Situ Secondary Self-Assembly of Near-Infrared II J-Aggregates: A Novel Phototheranostic Strategy for Inducing Tumor Pyroptosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2501184. [PMID: 40259472 DOI: 10.1002/adma.202501184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/07/2025] [Indexed: 04/23/2025]
Abstract
Pyroptosis, a programmed cell death mechanism that bypasses apoptosis resistance and triggers tumor-specific immune responses, has gained much attention as a promising approach to cancer therapy. Despite enhancing tumor accumulation and extending the circulation of small-molecule drugs, nanomedicines still face significant challenges, including poor tissue penetration, tumor resistance, and hypoxic microenvironments. To overcome these challenges, a novel near-infrared II (NIR-II) J-aggregate-based nanomedicine is designed, leveraging an in situ secondary self-assembly strategy to fabricate highly targeted nanoparticles (MSDP NPs). These nanomedicines trigger pyroptosis by generating type I reactive oxygen species, especially superoxide anions, while simultaneously activating photoimmunotherapy. In vivo studies demonstrate that MSDP NPs achieve efficient tumor penetration and prolong tumor retention, which is facilitated by the J-aggregate-driven formation of microscale spindle-shaped fibrillar bundles through in situ secondary self-assembly at the tumor site. This unique structural transformation enhances nanomedicine accumulation in tumor tissues, enabling robust NIR-II fluorescence imaging and improving therapeutic efficacy even in hypoxic tumor microenvironments. This study provides an innovative phototheranostic strategy that utilizes the in situ secondary self-assembly of NIR-II J-aggregates to induce tumor pyroptosis, offering a potential solution to the limitations of current nanomedicines in cancer therapy.
Collapse
Affiliation(s)
- Yuqi Tang
- Institute of Advanced Materials and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Dan Xiang
- Institute of Advanced Materials and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Quan Li
- Institute of Advanced Materials and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
- Materials Science Graduate Program, Kent State University, Kent, OH, 44242, USA
| |
Collapse
|
21
|
Zhu Y, Cai K, Guo L, Zhang H, Guan L, Zhang Z, Huang P, Yang S. Thrombospondin-1 induces immunogenic cell death in human mucoepidermoid carcinoma MC-3 cells via the PERK/eIF2α signaling pathway: potential implications for tumor immunotherapy. Discov Oncol 2025; 16:576. [PMID: 40253314 PMCID: PMC12009254 DOI: 10.1007/s12672-025-02315-7] [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: 12/02/2024] [Accepted: 04/04/2025] [Indexed: 04/21/2025] Open
Abstract
OBJECTIVE To investigate whether Thrombospondin-1 (TSP-1) induces immunogenic cell death (ICD) in human mucoepidermoid carcinoma (MC-3) cells and explore its potential to induce calreticulin (CRT) exposure via the PERK/eIF2α signaling pathway. METHODS The MC-3 cell line was used as the research model. The CCK-8 assay was performed to determine the optimal seeding density, TSP-1 concentration, and treatment time. Annexin V/PI double staining combined with flow cytometry was used to assess apoptosis across different experimental groups (blank control, TSP-1, paclitaxel (PTX), TSP-1 + PTX). Cells were divided into groups: blank control, PTX, TSP-1, TSP-1 + ISRIB (ISRIB: Integrated Stress Response Inhibitor), and TSP-1 + PTX, and CRT expression was detected by flow cytometry. Immunofluorescence, Western blot, and qPCR were used to detect the expression of PERK (Protein Kinase R-like Endoplasmic Reticulum Kinase), eIF2α (eukaryotic Initiation Factor 2α), and CRT. All experiments were performed in triplicate, and data were analyzed using GraphPad Prism 8.0 software. Statistical significance was set at P < 0.05. RESULTS At a seeding density of 2 × 104/mL, MC-3 cells reached the growth plateau by day six. The optimal concentration and duration of TSP-1 treatment were 0.1 μmol/L and 72 h, respectively. Flow cytometry, immunofluorescence, Western blot, and qPCR results revealed that TSP-1 significantly induced CRT exposure in MC-3 cells (P < 0.05), accompanied by the upregulation of PERK and eIF2α expression (P < 0.05). Co-treatment with PTX further enhanced these effects, while the addition of ISRIB reduced the expression of PERK, eIF2α, and CRT (P < 0.05). CONCLUSION TSP-1 induces ICD in MC-3 cells, accompanied by CRT exposure, potentially mediated through the activation of the PERK/eIF2α signaling pathway. These findings suggest that TSP-1 may have potential as an adjunct to chemotherapy for enhancing tumor immunotherapy.
Collapse
Affiliation(s)
- Yixuan Zhu
- North Sichuan Medical College, Nanchong, 637000, Sichuan, China
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining, Sichuan, China
| | - Kaizhi Cai
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining, Sichuan, China
| | - Lijuan Guo
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining, Sichuan, China
| | - Huan Zhang
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining, Sichuan, China
| | - Li Guan
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining, Sichuan, China
| | - Zongyao 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
| | - Pengcheng Huang
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining, Sichuan, China
| | - Sen Yang
- North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining, Sichuan, China.
| |
Collapse
|
22
|
Yang L, Wei W, Yuan X, Guo E, Peng P, Wang J, Sun W. Targeting DNA Damage Repair to Enhance Antitumor Immunity in Radiotherapy: Mechanisms and Opportunities. Int J Mol Sci 2025; 26:3743. [PMID: 40332379 PMCID: PMC12027993 DOI: 10.3390/ijms26083743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/13/2025] [Indexed: 05/08/2025] Open
Abstract
Radiotherapy is a standard cancer treatment that involves the induction of DNA damage. DNA damage repair (DDR) pathways maintain genomic integrity and make tumors resistant to radiotherapy and certain chemotherapies. In turn, DDR dysfunction results in cumulative DNA damage, leading to increased sensitivity for antitumor treatment. Moreover, radiotherapy has been shown to trigger antitumor immunity. Currently, immunotherapy has become a new and widely used standard strategy for treating a broad spectrum of tumor types. Notably, recent studies have demonstrated that DDR pathways play important roles in driving the response to immunotherapy. Herein, we review and discuss how DDR affects antitumor immunity induced by radiotherapy. Furthermore, we summarize the development of strategies for combining DDR inhibitors with radiotherapy and/or immunotherapy to enhance their efficacy against cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (L.Y.); (W.W.); (X.Y.); (E.G.); (P.P.); (J.W.)
| |
Collapse
|
23
|
Jinson S, Zhang Z, Lancaster GI, Murphy AJ, Morgan PK. Iron, lipid peroxidation, and ferroptosis play pathogenic roles in atherosclerosis. Cardiovasc Res 2025; 121:44-61. [PMID: 39739567 DOI: 10.1093/cvr/cvae270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Oxidation of lipids, excessive cell death, and iron deposition are prominent features of human atherosclerotic plaques. While extensive research has established the detrimental roles of lipid oxidation and apoptosis in atherosclerosis development, the involvement of iron in atherogenesis is not yet fully understood. With the emergence of an iron-dependent form of cell death termed ferroptosis, new attention has been brought to the complex inter-play among iron, ferroptosis, and atherosclerosis. Mechanistically, ferroptosis is caused by the lethal accumulation of iron-mediated lipid peroxides. Emerging studies have underscored ferroptosis as a contributor to worsened atherosclerosis. Herein, we review the evidence that oxidative damage and iron overload in the context of atherosclerosis may promote ferroptosis within plaques. Furthermore, we summarize recent findings of lipid peroxidation, thereby potentially ferroptosis, in various plaque cell types-such as endothelial cells, macrophages, dendritic cells, T cells, and vascular smooth muscle cells-across different stages of atherosclerosis. Understanding how these processes influence atherosclerotic plaque progression may permit targeting stage-dependent ferroptosis in each cell population and could provide a rationale for developing cell type-specific intervention strategies to mitigate atherogenic ferroptosis effectively.
Collapse
Affiliation(s)
- Swetha Jinson
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Ziyang Zhang
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Graeme I Lancaster
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Pooranee K Morgan
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| |
Collapse
|
24
|
Jiang J, Yan Y, Yang C, Cai H. Immunogenic Cell Death and Metabolic Reprogramming in Cancer: Mechanisms, Synergies, and Innovative Therapeutic Strategies. Biomedicines 2025; 13:950. [PMID: 40299564 PMCID: PMC12024911 DOI: 10.3390/biomedicines13040950] [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: 02/28/2025] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 05/01/2025] Open
Abstract
Immunogenic cell death (ICD) is a promising cancer therapy where dying tumor cells release damage-associated molecular patterns (DAMPs) to activate immune responses. Recent research highlights the critical role of metabolic reprogramming in tumor cells, including the Warburg effect, oxidative stress, and lipid metabolism, in modulating ICD and shaping the immune microenvironment. These metabolic changes enhance immune activation, making tumors more susceptible to immune surveillance. This review explores the molecular mechanisms linking ICD and metabolism, including mitochondrial oxidative stress, endoplasmic reticulum (ER) stress, and ferroptosis. It also discusses innovative therapeutic strategies, such as personalized combination therapies, metabolic inhibitors, and targeted delivery systems, to improve ICD efficacy. The future of cancer immunotherapy lies in integrating metabolic reprogramming and immune activation to overcome tumor immune evasion, with multi-omics approaches and microbiome modulation offering new avenues for enhanced treatment outcomes.
Collapse
Affiliation(s)
| | | | - Chunhui Yang
- Department of Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, China; (J.J.); (Y.Y.)
| | - Hong Cai
- Department of Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, China; (J.J.); (Y.Y.)
| |
Collapse
|
25
|
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
|
26
|
Lin B, Liu Y, Chen Q, Li M, Xu L, Chen Q, Tan Y, Liu Z. DNA Nanostructures-Based In Situ Cancer Vaccines: Mechanisms and Applications. SMALL METHODS 2025; 9:e2401501. [PMID: 39840607 DOI: 10.1002/smtd.202401501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/29/2024] [Indexed: 01/23/2025]
Abstract
Current tumor vaccines suffer from inadequate immune responsive due to the insufficient release of tumor antigens, low tumor infiltration, and immunosuppressive microenvironment. DNA nanostructures with their ability to precisely engineer, controlled release, biocompatibility, and the capability to augment the immunogenicity of tumor microenvironment, have gained significant attention for their potential to revolutionize vaccine designing. This review summarizes various applications of DNA nanostructures in the construction of in situ cancer vaccines, which can generate tumor-associated antigens directly from damaged tumors for cancer immune-stimulation. The mechanisms and components of cancer vaccines are listed, the specific strategies for constructing in situ vaccines using DNA nanostructures are explored and their underlying mechanisms of action are elucidated. The immunogenic cell death (ICD) induced by chemotherapeutic agents, photothermal therapy (PTT), photodynamic therapy (PDT), and radiation therapy (RT) and the related cancer vaccines building strategies are systematically summarized. The applications of different DNA nanostructures in various cancer immunotherapy are elaborated, which exerts precise, long-lasting, and robust immune responses. The current challenges and future prospectives are proposed. This review provides a holistic understanding of the evolving role of DNA nanostructures for in situ vaccine development.
Collapse
Affiliation(s)
- Bingyu Lin
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Qianqian Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| |
Collapse
|
27
|
Wang B, Zhang G, Chen Z, Shen H, Li C, Li J, Yi M, Sun J, Kwok RTK, Lam JWY, Qin A, Tang BZ. Lab-in-Cell: A Covalent Photosensitizer Reverses Hypoxia and Evokes Ferroptosis and Pyroptosis for Enhanced Anti-Tumor Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415673. [PMID: 39988860 DOI: 10.1002/adma.202415673] [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: 10/14/2024] [Revised: 02/12/2025] [Indexed: 02/25/2025]
Abstract
Photodynamic immunotherapy presents a non-invasive strategy characterized by spatiotemporal control and minimal side effects to induce immunogenic cell death (ICD). This approach significantly enhances the release of tumor-associated antigens and damage-associated molecular patterns, thereby improving cancer immunotherapy outcomes. However, hypoxia and antioxidant defenses at tumor sites considerably diminish the efficacy of photodynamic immunotherapy. In this work, a covalent warhead, alkyneamide, is introduced into an AIE photosensitizer to develop a novel covalent photosensitizer, MBTP-PA, which targets redox systems and facilitates ferroptosis- and pyroptosis-mediated photodynamic immunotherapy by thiol-yne click reactions. The covalent photosensitizer interacts with intracellular thiol compounds such as cysteine and glutathione, disrupting the intracellular antioxidant system and alleviating hypoxia. This results in enhanced photodynamic therapy (PDT) efficacy compared to the non-covalent photosensitizer MBTP-A. Furthermore, in conjunction with PDT, this reaction therapy can activate ICD through ferroptosis and pyroptosis, thereby enhancing anti-tumor immunity. Notably, in vivo injection of MBTP-PA nanoparticles at the tumor site led to the elimination of primary tumors, inhibiting distal tumors and exhibiting minimal side effects. Therefore, this work not only integrates the thiol-yne click reactions into cellular systems, significantly enhancing the efficacy of photodynamic immunotherapy but also paves the way for developing novel photosensitizers.
Collapse
Affiliation(s)
- Bingnan Wang
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, China
| | - Guiquan Zhang
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, Center for Aggregation-Induced Emission, AIE Institute, South China University of Technology, Guangzhou, 510640, China
| | - Zhizai Chen
- Department of Gynecology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Hanchen Shen
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, China
| | - Chunyang Li
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, Center for Aggregation-Induced Emission, AIE Institute, South China University of Technology, Guangzhou, 510640, China
| | - Jianqing Li
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, Center for Aggregation-Induced Emission, AIE Institute, South China University of Technology, Guangzhou, 510640, China
| | - Meixi Yi
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, Center for Aggregation-Induced Emission, AIE Institute, South China University of Technology, Guangzhou, 510640, China
| | - Jianwei Sun
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, China
| | - Ryan T K Kwok
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, China
| | - Jacky W Y Lam
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, China
| | - Anjun Qin
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, Center for Aggregation-Induced Emission, AIE Institute, South China University of Technology, Guangzhou, 510640, China
| | - Ben Zhong Tang
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, China
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, 518172, China
| |
Collapse
|
28
|
Jeong D, Kim S, Park H, Woo K, Choi J, Choi M, Shin J, Park SH, Seon M, Lee D, Cha J, Kim Y. Optogenetically Activatable MLKL as a Standalone Functional Module for Necroptosis and Therapeutic Applications in Antitumoral Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412393. [PMID: 39921454 PMCID: PMC11967802 DOI: 10.1002/advs.202412393] [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: 10/05/2024] [Revised: 01/17/2025] [Indexed: 02/10/2025]
Abstract
Necroptosis plays a crucial role in the progression of various diseases and has gained substantial attention for its potential to activate antitumor immunity. However, the complex signaling networks that regulate necroptosis have made it challenging to fully understand its mechanisms and translate this knowledge into therapeutic applications. To address these challenges, an optogenetically activatable necroptosis system is developed that allows for precise spatiotemporal control of key necroptosis regulators, bypassing complex upstream signaling processes. The system, specifically featuring optoMLKL, demonstrates that it can rapidly assemble into functional higher-order "hotspots" within cellular membrane compartments, independent of RIPK3-mediated phosphorylation. Moreover, the functional module of optoMLKL significantly enhances innate immune responses by promoting the release of iDAMPs and cDAMPs, which are critical for initiating antitumor immunity. Furthermore, optoMLKL exhibits antitumor effects when activated in patient-derived pancreatic cancer organoids, highlighting its potential for clinical application. These findings will pave the way for innovative cancer therapies by leveraging optogenetic approaches to precisely control and enhance necroptosis.
Collapse
Affiliation(s)
- Da‐Hye Jeong
- Department of BiochemistryAjou University School of MedicineSuwon16499Republic of Korea
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
| | - Seokhwi Kim
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
- Department of PathologyAjou University School of MedicineSuwon16499Republic of Korea
| | - Han‐Hee Park
- Department of BiochemistryAjou University School of MedicineSuwon16499Republic of Korea
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
| | - Kyoung‐Jin Woo
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
| | - Jae‐Il Choi
- Department of PathologyAjou University School of MedicineSuwon16499Republic of Korea
| | - Minji Choi
- Program in Biomedical Science and EngineeringGraduate schoolInha UniversityIncheon22212Republic of Korea
| | - Jisoo Shin
- Program in Biomedical Science and EngineeringGraduate schoolInha UniversityIncheon22212Republic of Korea
| | - So Hyun Park
- Department of PathologyAjou University School of MedicineSuwon16499Republic of Korea
| | - Myung‐Wook Seon
- Department of BiochemistryAjou University School of MedicineSuwon16499Republic of Korea
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
| | - Dakeun Lee
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
- Department of PathologyAjou University School of MedicineSuwon16499Republic of Korea
| | - Jong‐Ho Cha
- Program in Biomedical Science and EngineeringGraduate schoolInha UniversityIncheon22212Republic of Korea
- Department of Biomedical SciencesCollege of MedicineInha UniversityIncheon22212Republic of Korea
- Biohybrid Systems Research CenterInha UniversityIncheon22212Republic of Korea
| | - You‐Sun Kim
- Department of BiochemistryAjou University School of MedicineSuwon16499Republic of Korea
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
| |
Collapse
|
29
|
Tang M, Dirks K, Kim SY, Qiu Z, Gao Y, Sun D, Peruggia G, Sallavanti J, Li W. Inhibition of thioredoxin reductase 1 sensitizes glucose-starved glioblastoma cells to disulfidptosis. Cell Death Differ 2025; 32:598-612. [PMID: 39715824 PMCID: PMC11982235 DOI: 10.1038/s41418-024-01440-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024] Open
Abstract
Disulfidptosis is a recently identified form of cell death characterized by the aberrant accumulation of cellular disulfides. This process primarily occurs in glucose-starved cells expressing higher levels of SLC7A11 and has been proposed as a therapeutic strategy for cancers with hyperactive SCL7A11. However, the potential for inducing disulfidptosis through other mechanisms in cancers remains unclear. Here, we found that inhibiting thioredoxin reductase 1 (TrxR1), a key enzyme in the thioredoxin system, induces disulfidptosis in glioblastoma (GBM) cells. TrxR1 expression is elevated in GBM with activated transcriptional coactivator with PDZ-binding motif (TAZ) and correlates with poor prognosis. TrxR1 inhibitors induced GBM cell death that can be rescued by disulfide reducers but not by ROS scavengers or inhibitors of apoptosis, ferroptosis, or necroptosis. Glucose-starved cells, but not those deprived of oxygen or glutamine, increased TrxR1 expression in an NRF2-dependent manner and were more sensitive to TrxR1 inhibition-induced cell death. The dying cells initially exhibited highly dynamic lamellipodia, followed by actin cytoskeleton collapse. This process involved the accumulation of cytosolic peroxisomes and micropinocytic caveolae, as well as small gaps in the plasma membrane. Depletion of the WAVE complex component NCKAP1 partially rescued the cells, whereas Rac inhibition enhanced cell death. In an orthotopic xenograft GBM mouse model, TrxR1 depletion inhibited tumor growth and improved survival. Furthermore, cells undergoing TrxR1 inhibition exhibited features of immunogenic cell death. Therefore, this study suggests the potential of targeting TrxR1 as a therapeutic strategy in GBM.
Collapse
Affiliation(s)
- Miaolu Tang
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Kaitlyn Dirks
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
- Colorado State University, Fort Collins, USA
| | - Soo Yeon Kim
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Zhiqiang Qiu
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Yan Gao
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Dongxiao Sun
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Gabrielle Peruggia
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Jessica Sallavanti
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA.
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA.
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
30
|
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
|
31
|
Ye P, Wang C, Wen Y, Fang K, Li Q, Zhang X, Yang J, Li R, Chen M, Tong X, Shi S, Dong C. A positive-feedback loop suppresses TNBC tumour growth by remodeling tumour immune microenvironment and inducing ferroptosis. Biomaterials 2025; 315:122960. [PMID: 39541840 DOI: 10.1016/j.biomaterials.2024.122960] [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: 06/12/2024] [Revised: 09/10/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Triple-negative breast cancer (TNBC) is a particularly aggressive subtype of breast cancer due to poor immunogenicity and limited immune cell infiltration, efficient therapeutics are still deficiency. Ferroptosis, a reactive oxygen species (ROS)-reliant cell death, can enhance cellular immunogenicity and then active immune system. To sustain a long-term "hot" tumour immune microenvironment (TIME), an immune-modulator is indispensable. Metformin (MET), a commonly used oral drug for type 2 diabetes, has played a vital role in fostering an immunostimulatory environment. Herein, we confirm the TIME can be remodeled by MET and further promotes ferroptosis via upregulating cellular concentration of l-Glutamine. In light of this, we have design a self-assembled MET-loaded Fe3+-doped polydopamine nanoparticle (Fe-PDA-MET NP) that can disorder the cellular redox homeostasis and induce robust ferroptosis under 808 nm irradiation, resulting in a strong immune response. Based on the function of MET, there is a marked increase in the infiltration of activated CD8+ T cells and NK cells, which subsequently augments ferroptosis to a greater extent. Taken together, Fe-PDA-MET NPs activate a ferroptotic positive-feedback loop for effectively control TNBC progression, which offers a promising therapeutic modality to enhance the immunogenicity and reshape the TIME.
Collapse
Affiliation(s)
- Pingting Ye
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China
| | - Chunhui Wang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China
| | - Yixuan Wen
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China
| | - Kang Fang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China
| | - Qi Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China
| | - Xin Zhang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China
| | - Jingxian Yang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China
| | - Ruihao Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China
| | - Mengyao Chen
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China
| | - Xiaohan Tong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China
| | - Shuo Shi
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China.
| | - Chunyan Dong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
32
|
Macchia G, Pezzulla D, Campitelli M, Lucci S, Draghini L, Russo D, Fodor A, D'Agostino GR, Balcet V, Tamburo M, Giaccherini L, Tortoreto F, Augurio A, Ippolito E, Stefano AD, Fanelli M, Petrella L, Cilla S, Cosentino F, Marchetti C, Salutari V, Morganti AG, Gambacorta MA, Fagotti A, Pignata S, Scambia G, Ferrandina G, Deodato F. Treatment of Oligometastatic Parenchymal Lesions in Ovarian Cancer With Stereotactic Ablative Radiation Therapy: A Multicenter Prospective Phase 2 Trial (MITO RT3/RAD). Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00262-7. [PMID: 40174649 DOI: 10.1016/j.ijrobp.2025.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 04/04/2025]
Abstract
PURPOSE The results of stereotactic body radiation therapy (SBRT) for parenchymal lesions in the setting of oligometastatic ovarian cancer are reported in the context of the prospective multicenter phase 2 MITO-RT3/RAD trial (NCT04593381). METHODS AND MATERIALS The primary endpoint was the complete response (CR) rate, secondary endpoints included local control (LC), progression-free survival, overall survival, treatment-free interval, and toxicity rates. Sample size was based on a previous study reporting an average 40.0% CR with SBRT. The study was powered to detect an improvement in the CR rate from 40.0% to 55.0%, with an α error of 0.05 (one-side) and a β error of 0.1. RESULTS The study met its primary endpoint of a statistically significant improvement of CR. A total of 88 patients with 127 lesions were enrolled across 15 institutions from May 2019 to November 2023. CRs were observed in 71 lesions (55.9%), partial response in 37 (29.1%), stable disease in 14 (11.0%), and progressive disease in 5 lesions (4.0%). The objective response rate was 85.0%, with an overall clinical benefit rate of 96.0%. The overall 12-month LC was 81.6%, with CR lesions exhibiting a significantly higher rate than partial or not responding lesions (12-month LC: 96.3% vs 61.4%, P < .001). The 12-month actuarial rates for progression-free survival and for overall survival were 34.9% and 91.5%, respectively. The median actuarial treatment-free interval was 9 months (range, 2.5-15.4 months), whereas the 12-month actuarial rate was 44.1%. No grade 3 or higher toxicity was reported. In particular, 15 (20.5%) patients experienced mild acute toxicity (≤grade 2). There were 12 grade 1 events and 6 grade 2 events, the latter mostly represented by pain flare (N = 2). Late toxicity was reported in 4 patients (4.5%) accounting for 4 events, mostly grade 1, except for one case of moderate asthenia (grade 2). CONCLUSIONS Parenchymal oligometastatic lesions showed a high rate of CR and encouraging long-term outcomes for patients achieving CR, including a substantial period of systemic therapy-free survival after radiation therapy. The observed toxicity was minimal, strengthening the safety of ablative SBRT as a noninvasive alternative to surgical resection for parenchymal metastases in high-risk areas.
Collapse
Affiliation(s)
- Gabriella Macchia
- Radiation Oncology Unit, Responsible Research Hospital, Campobasso, Italy.
| | - Donato Pezzulla
- Radiation Oncology Unit, Responsible Research Hospital, Campobasso, Italy
| | - Maura Campitelli
- Unità Operativa Complessa di Radioterapia, Dipartimento di Scienze Radiologiche, Radioterapiche ed Ematologiche, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Roma, Italy
| | - Simona Lucci
- Unità Operativa Complessa di Radioterapia, Dipartimento di Scienze Radiologiche, Radioterapiche ed Ematologiche, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Roma, Italy
| | | | | | - Andrei Fodor
- Department of Radiation Oncology, Scientific Institute for Research, Hospitalization and Healthcare San Raffaele Scientific Institute, Milano, Italy
| | - Giuseppe Roberto D'Agostino
- Radiotherapy and Radiosurgery Department, Humanitas Clinical and Research Center-Scientific Institute for Research, Hospitalization and Healthcare, Milano, Italy
| | - Vittoria Balcet
- Unità Operativa Complessa Radioterapia, Nuovo Ospedale degli Infermi, Biella, Italy
| | - Marinella Tamburo
- Unità Operativa Complessa di Radioterapia, Azienda Ospedaliera Cannizzaro, Catania, Italy
| | - Lucia Giaccherini
- Radiation Oncology, Azienda USL - Istituto di Ricovero e Cura a Carattere Scientifico di Reggio Emilia, Reggio Emilia, Italy
| | - Francesca Tortoreto
- Unità Operativa Complessa di Radioterapia, Ospedale Isola Tiberina, Gemelli Isola, Rome, Italy
| | | | - Edy Ippolito
- Department of Radiation Oncology, Campus Bio-Medico University, Roma, Italy
| | - Aida Di Stefano
- Medical Oncology Unit, Responsible Research Hospital, Campobasso, Italy
| | - Mara Fanelli
- Research Laboratories, Responsible Research Hospital, Campobasso, Italy
| | - Lella Petrella
- Research Laboratories, Responsible Research Hospital, Campobasso, Italy
| | - Savino Cilla
- Medical Physics Unit, Responsible Research Hospital, Campobasso, Italy
| | - Francesco Cosentino
- Department of Gynecologic Oncology, Responsible Research Hospital, Campobasso, Italy; Università degli studi del Molise, Campobasso, Italy
| | - Claudia Marchetti
- Unità Operativa Complessa Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Roma, Italy
| | - Vanda Salutari
- Unità Operativa Complessa Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Roma, Italy
| | - Alessio Giuseppe Morganti
- Radiation Oncology, Istituto di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Radiation Oncology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Maria Antonietta Gambacorta
- Unità Operativa Complessa di Radioterapia, Dipartimento di Scienze Radiologiche, Radioterapiche ed Ematologiche, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Roma, Italy; Istituto di Radiologia, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Anna Fagotti
- Unità Operativa Complessa Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Roma, Italy
| | - Sandro Pignata
- Oncologia Clinica Sperimentale Uroginecologica Istituto Nazionale Tumori Istituto di Ricovero e Cura a Carattere Scientifico Fondazione G. Pascale, Napoli, Italy
| | - Giovanni Scambia
- Unità Operativa Complessa Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Roma, Italy
| | - Gabriella Ferrandina
- Unità Operativa Complessa Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Roma, Italy
| | - Francesco Deodato
- Radiation Oncology Unit, Responsible Research Hospital, Campobasso, Italy; Istituto di Radiologia, Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
33
|
Fang K, Yuan S, Zhang X, Zhang J, Sun SL, Li X. Regulation of immunogenic cell death and potential applications in cancer therapy. Front Immunol 2025; 16:1571212. [PMID: 40207233 PMCID: PMC11979251 DOI: 10.3389/fimmu.2025.1571212] [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: 02/05/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
Immunogenic cell death (ICD), a type of regulatory cell death, plays an important role in activating the adaptive immune response. Activation of the tumor-specific immune response is accompanied by the cell surface exposure of calreticulin and heat-shock proteins, the secretion of adenosine triphosphate, and the release of high mobility group box-1. In this review, we summarize and classify the latest types of ICD inducers and their molecular mechanisms, and discuss the effects and potential applications of inducing ICD by chemotherapy drugs, targeted drugs, and oncolytic viruses in clinical research. We also explore the potential role of epigenetic modifiers in the induction of ICD, and clarify the synergistic anti-tumor effects of nano-pulse stimulation, radiosensitizers for radiotherapy, photosensitizers for photodynamic therapy, photothermal therapy, and other physical stimulation, combined with radiotherapy and chemotherapy induced-ICD, in multimodal immunotherapy. In addition, we elucidate the molecular mechanism of ICD in detail, including the calcium imbalance, mitochondrial stress, and the interactions in the tumor microenvironment. Ultimately, this review aims to offer deeper insight into the factors and mechanisms of ICD induction and provide a theoretical basis for the future development of ICD-based immunotherapy.
Collapse
Affiliation(s)
- Kun Fang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Shuai Yuan
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Xue Zhang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Jingdong Zhang
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
- Department of Medical Oncology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
| | - Shu-lan Sun
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Xiaoxi Li
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| |
Collapse
|
34
|
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
|
35
|
Singh S, Kim GH, Baek KR, Seo SO. Anti-Cancer Strategies Using Anaerobic Spore-Forming Bacteria Clostridium: Advances and Synergistic Approaches. Life (Basel) 2025; 15:465. [PMID: 40141809 PMCID: PMC11943571 DOI: 10.3390/life15030465] [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: 02/25/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Despite ongoing advancements, cancer remains a significant global health concern, with a persistent challenge in identifying a definitive cure. While various cancer therapies have been developed and approved, offering treatments for smaller neoplasms, their efficacy diminishes in solid tumors and hypoxic environments, particularly for chemotherapy and radiation therapy. A novel approach, Clostridium-based therapy, has emerged as a promising candidate for current solid tumor treatments due to its unique affinity for the hypoxic tumor microenvironment. This review examines the potential of Clostridium in cancer treatment, encompassing direct tumor lysis, immune modulation, and synergistic effects with existing cancer therapies. Advancements in synthetic biology have further enhanced its potential through genetic modifications, such as the removal of alpha toxin gene from Clostridium novyi-NT, the implementation of targeted approaches, and reduction in systemic toxicity. Although preclinical and clinical studies have demonstrated that Clostridium-based treatments combined with other therapies hold promise for complete cancer eradication, challenges persist. Through this review, we also propose that the integration of various methods and technologies together with Clostridium-based therapy may lead to the complete eradication of cancer in the future.
Collapse
Affiliation(s)
- Saloni Singh
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea (G.-H.K.); (K.-R.B.)
| | - Geun-Hyung Kim
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea (G.-H.K.); (K.-R.B.)
| | - Kwang-Rim Baek
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea (G.-H.K.); (K.-R.B.)
| | - Seung-Oh Seo
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea (G.-H.K.); (K.-R.B.)
- Research Institute of Food and Biotechnology, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| |
Collapse
|
36
|
De Robertis M, Bozic T, Santek I, Marzano F, Markelc B, Silvestris DA, Tullo A, Pesole G, Cemazar M, Signori E. Transcriptomic analysis of the immune response to in vivo gene electrotransfer in colorectal cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102448. [PMID: 39967849 PMCID: PMC11834060 DOI: 10.1016/j.omtn.2025.102448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025]
Abstract
Gene electrotransfer (GET) has recently emerged as an effective nonviral approach for plasmid DNA (pDNA) delivery in gene therapy for several pathologies, including cancer. Multiple mechanisms have been identified that influence cell biology after GET, as electroporation significantly increases pDNA uptake and immunogenicity, which may directly influence target cell death. However, the molecular effects of in vivo electroporation-mediated DNA delivery have yet to be fully elucidated. In this study, we evaluated the transcriptomes of murine colorectal tumors treated with two protocols, short- and high-voltage (SHV) electric pulses or an adapted high-voltage-low-voltage (HV-LV) pulse protocol, both of which are used for reversible electroporation. Although no significant differences in clinical outcomes were observed, variations in intratumoral macrophage infiltration were reported between the two treatment methods. Transcriptomic analysis revealed that apoptosis is a predominant mode of cell death after GET by SHV pulses, whereas GET by HV-LV pulses is associated with immunogenic necrotic pathways as well as the activation of both the innate and adaptive immune response. We demonstrated that specific pulse parameters can induce distinct immunomodulatory profiles in the tumor microenvironment; therefore, these aspects should be considered carefully when selecting the most suitable GET-based approach for antitumor immunization.
Collapse
Affiliation(s)
- Mariangela De Robertis
- Department of Biosciences, Biotechnology, and Environment, University of Bari “Aldo Moro”, 70126 Bari, Italy
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Tim Bozic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
| | - Iva Santek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Flaviana Marzano
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Bostjan Markelc
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva ulica 101, 1000 Ljubljana, Slovenia
| | | | - Apollonia Tullo
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Graziano Pesole
- Department of Biosciences, Biotechnology, and Environment, University of Bari “Aldo Moro”, 70126 Bari, Italy
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, Polje 42, 6310 Izola, Slovenia
| | - Emanuela Signori
- Laboratory of Molecular Pathology and Experimental Oncology, Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche, 0133 Rome, Italy
| |
Collapse
|
37
|
Yan Z, Bai Y, Zhang S, Kong L, Wang Y, Sun H, Li Y, Qiu L, Zhang R, Jiang P, Zhao D, Chen Z, Li Y, Pang H, Wang J. Quasi Fe MIL-53 nanozyme inducing ferroptosis and immunogenic cell death for cancer immunotherapy. Nat Commun 2025; 16:2290. [PMID: 40055308 PMCID: PMC11889140 DOI: 10.1038/s41467-025-57542-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
Nanozymes offer diverse therapeutic potentials for cancer treatment which is dependent on the development of nanomaterials. Quasi-metal-organic framework is a class of metal-organic framework-derived nanomaterials with a transition state from metal-organic frameworks towards metal oxide featuring porous structure and high activity. Herein an iron-based quasi-metal-organic framework nanozyme Q-MIL-53(Fe) is reported via a controlled deligandation strategy, exhibiting enhanced peroxidase-/catalase-mimic activity and glutathione depletion capacity, whose underlying mechanisms are studied via density functional theory calculations. Q-MIL-53(Fe) demonstrates biocompatibility and superior antitumor efficacy compared to pristine MIL-53(Fe). It can activate antitumor immune response by inducing ferroptosis and immunogenic cell death, promoting dendritic cell maturation and T lymphocytes infiltration. Furthermore, a combination of Q-MIL-53(Fe) and programmed cell death protein 1 antibody amplifies cancer immunotherapy. This study validates the antitumor activity of quasi-metal-organic frameworks and its immunotherapy induction potential. It would broaden the application of quasi-metal-organic frameworks and open avenues for developing antitumor nanozymes.
Collapse
Affiliation(s)
- Zihui Yan
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Yang Bai
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China.
| | - Songtao Zhang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225002, P. R. China
| | - Lingyi Kong
- Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Yu Wang
- Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Huilin Sun
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Yi Li
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Ruijie Zhang
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Pengju Jiang
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Donghui Zhao
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Zhongyan Chen
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, 325035, P. R. China
| | - Yafei Li
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China.
- Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China.
| | - Huan Pang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225002, P. R. China.
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China.
- School of Medical and Health Engineering, Changzhou University, Changzhou, 213164, P. R. China.
| |
Collapse
|
38
|
Sun S, Qian S, Wang R, Zhao M, Li R, Gu W, Zhao M, Qian C, Liu L, Tang X, Li Y, Shi H, Pan Y, Xiao H, Yang K, Hu C, Huang Y, Wei L, Zhang Y, Ji J, Chen Y, Liu H. Targeting GOLPH3L improves glioblastoma radiotherapy by regulating STING-NLRP3-mediated tumor immune microenvironment reprogramming. Sci Transl Med 2025; 17:eado0020. [PMID: 40043140 DOI: 10.1126/scitranslmed.ado0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 08/04/2024] [Accepted: 02/12/2025] [Indexed: 04/18/2025]
Abstract
Radiotherapy (RT) has been the standard-of-care treatment for patients with glioblastoma (GBM); however, the clinical effectiveness is hindered by therapeutic resistance. Here, we demonstrated that the tumor immune microenvironment (TIME) exhibited immunosuppressive properties and high expression of Golgi phosphoprotein 3 like (GOLPH3L) in RT-resistant GBM. Our study showed that GOLPH3L interacted with stimulator of interferon genes (STING) at the aspartic acid residue 184 in Golgi after RT, leading to coat protein complex II-mediated retrograde transport of STING from Golgi to endoplasmic reticulum. This suppressed the STING-NOD-like receptor thermal protein domain associated protein 3 (NLRP3)-mediated pyroptosis, resulting in suppressive TIME, driving GBM resistance to RT. Genetic GOLPH3L ablation in RT-resistant GBM cells augmented antitumor immunity and overcame tumor resistance to RT. Moreover, we have identified a small molecular inhibitor of GOLPH3L, vitamin B5 calcium (VB5), which improved the therapeutic efficacy of RT and immune checkpoint blockade by inducing a robust antitumor immune response in mouse models. Clinically, patients with GBM treated with VB5 exhibited improved responses to RT. Thus, reprogramming the TIME by targeting GOLPH3L may offer a potential opportunity to improve RT in GBM.
Collapse
Affiliation(s)
- Shuo Sun
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shiyu Qian
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Ran Wang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mengya Zhao
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
- Research Center of Surgery, Nanjing BenQ Medical Center, Affiliated BenQ Hospital of Nanjing Medical University, Department of Immunology, Nanjing Medical University, Nanjing 211166, China
| | - Ran Li
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Wei Gu
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mengjie Zhao
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chunfa Qian
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xianglong Tang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yangyang Li
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hui Shi
- Department of Neurosurgery, First Hospital of Lianyungang, Lianyungang 222000, China
| | - Yunsong Pan
- Department of Neurosurgery, First Hospital of Lianyungang, Lianyungang 222000, China
| | - Hong Xiao
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Kun Yang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chupeng Hu
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Yedi Huang
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Liangnian Wei
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Yuhan Zhang
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Jing Ji
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yun Chen
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Hongyi Liu
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
39
|
Chang MR, Matnurov EM, Wu C, Arakelyan J, Choe HJ, Kushnarev V, Yap JY, Soo XX, Chow MJ, Berger W, Ang WH, Babak MV. Leveraging Immunogenic Cell Death to Enhance the Immune Response against Malignant Pleural Mesothelioma Tumors. J Am Chem Soc 2025; 147:7908-7920. [PMID: 39992709 PMCID: PMC11887451 DOI: 10.1021/jacs.4c17966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025]
Abstract
Although various metal-based compounds have exhibited excellent immunogenic cell death (ICD)-inducing properties both in vitro and in vivo, the majority of these compounds have been discovered serendipitously. In this work, we have successfully synthesized and characterized 35 cyclometalated Au(III) complexes containing dithiocarbamate ligands, with 25 of these complexes being previously unreported. Their ability to induce phagocytosis in vitro against immunologically "cold" malignant pleural mesothelioma (MPM) cells was strongly dependent on the cyclometalated scaffold and the overall lipophilicity of the complexes. We elucidated the role of cell death mechanisms in the observed ICD effects and identified correlations between the ability of the complexes to induce necrotic cell death and ICD, both in vitro and in vivo. Complex 2G, with its high phagocytosis rates and low necrosis rates, was recognized as a bona fide ICD inducer, demonstrating a remarkably long-lasting immune response in vaccinated mice. In contrast, complex 1C, characterized by high phagocytosis rates and high necrosis rates, failed to elicit a sustained immune response upon following vaccination; however, it triggered selective activation of calreticulin in tumors upon direct in vivo administration. Overall, this study offers a framework for predicting ICD effects in vivo for structurally similar Au(III) complexes, with the potential for extension to other series of metal complexes.
Collapse
Affiliation(s)
- Meng Rui Chang
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Egor M. Matnurov
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Chengnan Wu
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Jemma Arakelyan
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Ho-Jung Choe
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Vladimir Kushnarev
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Jian Yu Yap
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
- NUS Graduate
School - Integrated Science and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Xiu Xuan Soo
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Mun Juinn Chow
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Walter Berger
- Center for
Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, Vienna 1090, Austria
| | - Wee Han Ang
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
- NUS Graduate
School - Integrated Science and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, 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
| |
Collapse
|
40
|
Xiong Y, Sun M, Yang Q, Zhang W, Song A, Tan Y, Mao J, Liu G, Xue P. Nanoparticle-based drug delivery systems to modulate tumor immune response for glioblastoma treatment. Acta Biomater 2025; 194:38-57. [PMID: 39884522 DOI: 10.1016/j.actbio.2025.01.050] [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: 10/16/2024] [Revised: 12/28/2024] [Accepted: 01/28/2025] [Indexed: 02/01/2025]
Abstract
Glioblastoma (GBM) is a primary central nervous system neoplasm, characterized by a grim prognosis and low survival rates. This unfavorable therapeutic outcome is partially attributed to the inadequate immune infiltration and an immunosuppressive microenvironment, which compromises the effectiveness of conventional radiotherapy and chemotherapy. To this end, precise modulation of cellular dynamics in the immune system has emerged as a promising approach for therapeutic intervention. The advent of nanoparticle-based therapies has revolutionized cancer treatment and provided highly effective options. Consequently, various strategically designed nano-delivery platforms have been established to promote the efficacy of immune therapy against GBM. This review delves into the recent advancements in nano-based delivery systems that are designed to modulate immune cells in GBM microenvironment, and explores their multifaceted mechanisms, including the blockade of immune checkpoints, the restraint of immunosuppressive cells, the coordination of tumor-associated macrophages, the activation of innate immune cells, and the stimulation of adaptive immunity. Collectively, this summary not only advances the comprehension involved in modulating antitumor immune responses in GBM, but also paves the way for the development of innovative therapeutic strategies to conquer GBM. STATEMENT OF SIGNIFICANCE: Glioblastoma (GBM) is the most lethal brain tumor, with a median survival rate of merely 12-16 months after diagnosis. Despite surgical, radiation and chemotherapy treatments, the two-year survival rate for GBM patients is less than 10 %. The treatment of GBM is challenging mainly because several issues associated with the GBM microenvironment have not yet been resolved. Most recently, novel drug delivery approaches, based on the clear understanding of the intrinsic properties of GBM, have shown promise in overcoming some of the obstacles. In particular, taking account of the highly immunosuppressive tumor microenvironment in GBM, recent advancements in nano-based delivery systems are put forward to stimulate immune cells in GBM and unravel their multifaceted mechanisms. This review summarizes the latest nanoparticle-based drug delivery systems to modulate tumor immune response for glioblastoma treatment. Moreover, the development trends and challenges of nanoparticle-based drug delivery systems in modulating the immunity of GBM are predicted, which may facilitate widespread regimens springing up for successfully treating GBM.
Collapse
Affiliation(s)
- Yongqi Xiong
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Maoyuan Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qinhao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wenli Zhang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Anchao Song
- College of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Ying Tan
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang 550000, China
| | - Jinning Mao
- Health Medical Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Peng Xue
- School of Materials and Energy, Southwest University, Chongqing 400715, China; Yibin Academy of Southwest University, Yibin 644005, China.
| |
Collapse
|
41
|
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
|
42
|
Morris ZS, Demaria S, Monjazeb AM, Formenti SC, Weichselbaum RR, Welsh J, Enderling H, Schoenfeld JD, Brody JD, McGee HM, Mondini M, Kent MS, Young KH, Galluzzi L, Karam SD, Theelen WSME, Chang JY, Huynh MA, Daib A, Pitroda S, Chung C, Serre R, Grassberger C, Deng J, Sodji QH, Nguyen AT, Patel RB, Krebs S, Kalbasi A, Kerr C, Vanpouille-Box C, Vick L, Aguilera TA, Ong IM, Herrera F, Menon H, Smart D, Ahmed J, Gartrell RD, Roland CL, Fekrmandi F, Chakraborty B, Bent EH, Berg TJ, Hutson A, Khleif S, Sikora AG, Fong L. Proceedings of the National Cancer Institute Workshop on combining immunotherapy with radiotherapy: challenges and opportunities for clinical translation. Lancet Oncol 2025; 26:e152-e170. [PMID: 40049206 DOI: 10.1016/s1470-2045(24)00656-9] [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: 08/07/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 03/09/2025]
Abstract
Radiotherapy both promotes and antagonises tumour immune recognition. Some clinical studies show improved patient outcomes when immunotherapies are integrated with radiotherapy. Safe, greater than additive, clinical response to the combination is limited to a subset of patients, however, and how radiotherapy can best be combined with immunotherapies remains unclear. The National Cancer Institute-Immuno-Oncology Translational Network-Society for Immunotherapy of Cancer-American Association of Immunology Workshop on Combining Immunotherapy with Radiotherapy was convened to identify and prioritise opportunities and challenges for radiotherapy and immunotherapy combinations. Sessions examined the immune effects of radiation, barriers to anti-tumour immune response, previous clinical trial data, immunological and computational assessment of response, and next-generation radiotherapy-immunotherapy combinations. Panel recommendations included: developing and implementing patient selection and biomarker-guided approaches; applying mechanistic understanding to optimise delivery of radiotherapy and selection of immunotherapies; using rigorous preclinical models including companion animal studies; embracing data sharing and standardisation, advanced modelling, and multidisciplinary cross-institution collaboration; interrogating clinical data, including negative trials; and incorporating novel clinical endpoints and trial designs.
Collapse
Affiliation(s)
- Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Sandra Demaria
- Weill Cornell Medicine, Department of Radiation Oncology, New York, NY, USA
| | - Arta M Monjazeb
- UC Davis Health, Department of Radiation Oncology, Sacramento, CA, USA
| | - Silvia C Formenti
- Weill Cornell Medicine, Department of Radiation Oncology, New York, NY, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA
| | - James Welsh
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Heiko Enderling
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Joshua D Brody
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Heather M McGee
- Department of Radiation Oncology and Department of Immuno-Oncology, City of Hope, Duarte, CA, USA
| | - Michele Mondini
- Gustave Roussy, Université Paris-Saclay, INSERM U1030, Villejuif, France
| | - Michael S Kent
- Davis School of Veterinary Medicine, University of California, Davis, CA, USA
| | | | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Joe Y Chang
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Mai Anh Huynh
- Brigham and Women's Hospital-Dana-Farber Cancer Institute, Boston, MA, USA
| | - Adi Daib
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Sean Pitroda
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA
| | - Caroline Chung
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Raphael Serre
- Aix Marseille University, SMARTc Unit, Inserm S 911 CRO2, Marseille, France
| | | | - Jie Deng
- Department of Radiation Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Quaovi H Sodji
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Anthony T Nguyen
- Cedars-Sinai Medical Center, Department of Radiation Oncology, Los Angeles, CA, USA
| | - Ravi B Patel
- Department of Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA, USA
| | - Simone Krebs
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medicine, Department of Radiology, New York, NY, USA
| | - Anusha Kalbasi
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, USA
| | - Caroline Kerr
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Logan Vick
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | | | - Irene M Ong
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Fernanda Herrera
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland
| | - Hari Menon
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - DeeDee Smart
- Radiation Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Jalal Ahmed
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robyn D Gartrell
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA; Department of Oncology, Division of Pediatric Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Christina L Roland
- Department of Thoracic Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Fatemeh Fekrmandi
- Department of Radiation Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Binita Chakraborty
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Eric H Bent
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tracy J Berg
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Alan Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Samir Khleif
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Andrew G Sikora
- Department of Head and Neck Surgery, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Lawrence Fong
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| |
Collapse
|
43
|
Collins VG, Hutton D, Hossain-Ibrahim K, Joseph J, Banerjee S. The abscopal effects of sonodynamic therapy in cancer. Br J Cancer 2025; 132:409-420. [PMID: 39537767 PMCID: PMC11876350 DOI: 10.1038/s41416-024-02898-y] [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: 07/21/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The abscopal effect is a phenomenon wherein localised therapy on the primary tumour leads to regression of distal metastatic growths. Interestingly, various pre-clinical studies utilising sonodynamic therapy (SDT) have reported significant abscopal effects, however, the mechanism remains largely enigmatic. SDT is an emerging non-invasive cancer treatment that uses focussed ultrasound (FUS) and a sonosensitiser to induce tumour cell death. To expand our understanding of abscopal effects of SDT, we have summarised the preclinical studies that have found SDT-induced abscopal responses across various cancer models, using diverse combination strategies with nanomaterials, microbubbles, chemotherapy, and immune checkpoint inhibitors. Additionally, we shed light on the molecular and immunological mechanisms underpinning SDT-induced primary and metastatic tumour cell death, as well as the role and efficacy of different sonosensitisers. Notably, the observed abscopal effects underscore the need for continued investigation into the SDT-induced 'vaccine-effect' as a potential strategy for enhancing systemic anti-tumour immunity and combating metastatic disease. The results of the first SDT human clinical trials are much awaited and are hoped to enable the further evaluation of the safety and efficacy of SDT, paving the way for future studies specifically designed to explore the potential of translating SDT-induced abscopal effects into clinical reality.
Collapse
Affiliation(s)
- Victoria G Collins
- Department of Neurosurgery, Ninewells Hospital, Dundee, UK
- Department of Neurosurgery, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Dana Hutton
- The Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | | | - James Joseph
- Department of Biomedical Engineering, School of Science and Engineering, University of Dundee, Dundee, UK.
| | - Sourav Banerjee
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee, UK.
| |
Collapse
|
44
|
Macchia G, Campitelli M, Pezzulla D, Lucci S, Fodor A, Russo D, Balcet V, Bonome P, Durante S, Draghini L, Titone F, D'Agostino GR, Tamburo M, Ferioli M, Ippolito E, Tortoreto F, Caravatta L, De Felice F, Stefano AD, Fanelli M, Cilla S, Cosentino F, Marchetti C, Salutari V, Boccia S, Morganti AG, Gambacorta MA, Fagotti A, Pignata S, Scambia G, Ferrandina G, Deodato F. Stereotactic Ablative Radiation Therapy for Oligometastatic Ovarian Cancer Lymph Node Disease: The MITO-RT3/RAD Phase II Trial. Int J Radiat Oncol Biol Phys 2025; 121:693-702. [PMID: 39326506 DOI: 10.1016/j.ijrobp.2024.09.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/05/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024]
Abstract
PURPOSE MITO-RT3/RAD (NCT04593381) is a prospective multicenter phase 2 trial designed to assess the effectiveness and safety of stereotactic body radiation therapy (SBRT) in patients who received diagnoses of oligometastatic ovarian cancer. In this report, we provide the results of the trial in the setting of lymph node disease. METHODS AND MATERIALS The primary endpoint was the complete response (CR) rate, secondary endpoints included local control (LC), progression-free survival (PFS), overall survival, treatment-free interval, and toxicity rates. The sample size was based on a previous study reporting an average 70.0% CR with SBRT. The study was powered to detect an improvement in the CR rate from 70.0% to 85.0%, with an α error of 0.05 (one-side) and a β error of 0.1. RESULTS The study met its primary endpoint of a statistically significant improvement in CR. One hundred thirty-five patients with 249 lesions were enrolled across 15 institutions from May 2019 to November 2023. CRs were observed in 194 lesions (77.9%), partial responses in 40 (16.1%), stable disease in 14 (5.6%), and progressive disease in 1 lesion (0.4%). The objective response rate was 94%, with an overall clinical benefit rate of 99.6%. CR lesions exhibited a significantly higher LC rate than partial or not responding lesions (12-month LC: 92.7% vs 63.1%, P < .001). The 12-month actuarial rates for PFS and for overall survival were 36.6% (CR, 38.3% vs not-CR, 18.8%; P, .022) and 97.2% (CR, 97.8% vs not-CR, 93.8%; P, .067), respectively. The 12-month actuarial rate for treatment-free interval was 52.7% (CR, 58.4% vs not-CR, 24.4%; P, .004). CR was substantially associated with higher PFS (P, .036) and treatment-free interval (P, .006) rates in the univariate analysis. Twenty-three patients (17.0%) experienced mild acute toxicity. Late toxicity was reported in 9 patients (6.7%), mostly grade 1. CONCLUSIONS This trial confirms the efficacy of ablative SBRT, with minimal toxicity observed. SBRT offered a high CR rate, promising long-term outcomes, and a significant systemic therapy-free survival period for complete responders.
Collapse
Affiliation(s)
- Gabriella Macchia
- Radiation Oncology Unit, Responsible Research Hospital, Campobasso, Molise, Italy.
| | - Maura Campitelli
- UOC di Radioterapia, Dipartimento di Scienze Radiologiche, Radioterapiche ed Ematologiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Donato Pezzulla
- Radiation Oncology Unit, Responsible Research Hospital, Campobasso, Molise, Italy
| | - Simona Lucci
- UOC di Radioterapia, Dipartimento di Scienze Radiologiche, Radioterapiche ed Ematologiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Andrei Fodor
- Department of Radiation Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Vittoria Balcet
- UOC Radioterapia, Nuovo Ospedale degli Infermi, Biella, Italy
| | - Paolo Bonome
- Radiation Oncology Unit, Responsible Research Hospital, Campobasso, Molise, Italy
| | - Stefano Durante
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, and Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | | | - Francesca Titone
- Department of Radiation Oncology, University Hospital Udine, Italy
| | - Giuseppe Roberto D'Agostino
- Radiotherapy and Radiosurgery Department, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Marinella Tamburo
- U.O.C. di Radioterapia, Azienda Ospedaliera "Cannizzaro" - Catania, Italy
| | - Martina Ferioli
- Radiation Oncology, Azienda USL - IRCCS di Reggio Emilia, Italy
| | - Edy Ippolito
- Department of Radiation Oncology, Campus Bio-Medico University, Roma, Italy
| | - Francesca Tortoreto
- UOC di Radioterapia Fatebenefratelli Isola Tiberina. Gemelli Isola, Rome, Italy
| | - Luciana Caravatta
- Department of Radiation Oncology, SS Annunziata Hospital, Chieti, Italy
| | - Francesca De Felice
- Department of Radiotherapy, Policlinico Umberto I, Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Aida Di Stefano
- Medical Oncology Unit, Responsible Research Hospital, Campobasso, Molise, Italy
| | - Mara Fanelli
- Research Laboratories, Responsible Research Hospital, Campobasso, Molise, Italy
| | - Savino Cilla
- Medical Physics Unit, Responsible Research Hospital, Campobasso, Molise, Italy
| | - Francesco Cosentino
- Department of Gynecologic Oncology, Responsible Research Hospital and Università degli studi del Molise, Campobasso, Italy
| | - Claudia Marchetti
- UOC Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Vanda Salutari
- UOC Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Serena Boccia
- UOC Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Alessio Giuseppe Morganti
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna; Radiation Oncology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Maria Antonietta Gambacorta
- UOC di Radioterapia, Dipartimento di Scienze Radiologiche, Radioterapiche ed Ematologiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy; Istituto di Radiologia, Università Cattolica del Sacro Cuore Roma, Italy
| | - Anna Fagotti
- UOC Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Sandro Pignata
- Oncologia Clinica Sperimentale Uroginecologica Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Giovanni Scambia
- UOC Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Gabriella Ferrandina
- UOC Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Francesco Deodato
- Radiation Oncology Unit, Responsible Research Hospital, Campobasso, Molise, Italy; Istituto di Radiologia, Università Cattolica del Sacro Cuore Roma, Italy
| |
Collapse
|
45
|
Yang Z, Zhou Y, Liu X, Ren L, Liu X, Yun R, Jia L, Ren X, Wang Y, Sun Y, Li J, Gao D, Tian Z. Mitochondrial-uncoupling nanomedicine for self-heating and immunometabolism regulation in cancer cells. Biomaterials 2025; 314:122883. [PMID: 39405827 DOI: 10.1016/j.biomaterials.2024.122883] [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: 07/25/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 11/10/2024]
Abstract
Developing endogenous hyperthermia offers a promising strategy to address challenges with current exogenous hyperthermia techniques in clinics. Herein, a CD44-targeted and thermal-responsive nanocarrier was developed for the simultaneous delivery of 2,4-dinitrophenol and syrosingopine. The objective was to induce endogenous hyperthermia and regulate immunometabolism, ultimately augmenting anti-tumour immune responses. Dinitrophenol as mitochondrial uncoupler can convert electrochemical potential energy of inner mitochondrial membrane into heat, facilitating endogenous hyperthermia. Meanwhile, syrosingopine not only inhibits excessive lactate efflux caused by dinitrophenol but also downregulates tumour cell glycolysis, thus alleviating immunosuppression and heat shock protein (HSP)-dependent thermo-resistance through immunometabolism regulation. The synergistic effects of endogenous hyperthermia and immunometabolism regulation by this nanomedicine have potential to enhance tumor immunogenicity, reshape the tumour immune microenvironment, and effectively suppress the growth of subcutaneous tumours and patient-derived organoids in triple-negative breast cancer. Therefore, the endogenous hyperthermia strategy developed in this study would revolutionize hyperthermia for cancer treatment.
Collapse
Affiliation(s)
- Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Ying Zhou
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiaozhen Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; General Surgery, Department of Breast Surgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Liujiao Ren
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinyang Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Rong Yun
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Liangliang Jia
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xuechun Ren
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ying Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yan Sun
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jia Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Di Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
46
|
Zheng L, Wang H, Zhong X, Jia L, Shi G, Bai C, Yang R, Huang Z, Jiang Y, Wei J, Dong Z, Li J, Long Y, Dai L, Li Z, Chen C, Wang J. Reprogramming tumor microenvironment with precise photothermal therapy by calreticulin nanobody-engineered probiotics. Biomaterials 2025; 314:122809. [PMID: 39303415 DOI: 10.1016/j.biomaterials.2024.122809] [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: 03/31/2024] [Revised: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024]
Abstract
Targeted therapies have revolutionized traditional cancer treatments by precisely targeting tumor cells, enhancing efficacy and safety. Despite this advancement, the proportion of cancer patients eligible for such therapies remains low due to the absence of suitable targets. Here, we investigate whether the translocation of the immunogenic cell death (ICD) marker calreticulin (CALR) from the endoplasmic reticulum (ER) to the cell surface following ICD induction can serve as a target for targeted therapies. To target CALR, a nanobody Nb215 identified from a naïve VHH phage library with high binding affinity to both human and mouse CALR was employed to engineer probiotic EcN 1917. Our results demonstrated that CALR nanobody-modified EcN-215 coupled with the photothermal dye indocyanine green (ICG) was able to exert NIR-II imaging-guide photothermal therapy (PTT). Moreover, PTT with EcN-215/ICG can reshape the tumor microenvironment by enhancing the infiltration of CD45+CD3+ T cells and CD11b+F4/80+ macrophages. Furthermore, the antitumor activity of CALR-targeted EcN-215/ICG is synergistically enhanced by blocking CD47-SIRPα axis. Collectively, our study provides a proof of concept for CALR-targeted therapy. Given that CALR translocation can be induced by various anticancer therapies across numerous tumor cell lines, CALR-targeted therapies hold promise as a novel approach for treating multiple types of cancers.
Collapse
Affiliation(s)
- Liuhai Zheng
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Huifang Wang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xiaoru Zhong
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Lin Jia
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Guangwei Shi
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528399, China
| | - Chongzhi Bai
- Central Laboratory, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, 030012, China
| | - Runwei Yang
- Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528399, China
| | - Zhenhui Huang
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Yuke Jiang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China; State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Jinxi Wei
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Zhiyu Dong
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Jiexuan Li
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Ying Long
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Lingyun Dai
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China.
| | - Zhijie Li
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China.
| | - Chunbo Chen
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China.
| | - Jigang Wang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China; State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
47
|
Chen CC, Hsu LW, Chen KD, Chiu KW, Kung CP, Li SR, Goto S, Chen CL, Huang KT. Extracellular calreticulin regulates fibrogenic and immunogenic properties of hepatic stellate cells. Int Immunopharmacol 2025; 148:114129. [PMID: 39862632 DOI: 10.1016/j.intimp.2025.114129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Liver fibrosis is a persistent damage repair response triggered by various etiological factors, resulting in an excessive accumulation of extracellular matrix (ECM). Activated hepatic stellate cells (HpSCs) are the primary source of ECM proteins. Therefore, specifically targeting HpSCs has become a crucial approach for treating liver fibrosis. Calreticulin (CRT) is a molecular chaperone mainly located in the endoplasmic reticulum (ER), regulating protein folding and calcium homeostasis. Recently, CRT has gained much attention for its role outside the ER, particularly at the cell surface and extracellular space, acting as an immunomodulatory protein. The current study investigates the role of extracellular CRT in hepatic injury and its effects on HpSCs. Elevated levels of circulating CRT were observed in mouse models of liver injury, suggesting that hepatic injury may trigger CRT release. Extracellular CRT was found to moderately inhibit HpSC viability and induce morphological changes. Additionally, CRT treatment led to a decrease in α-smooth muscle actin and an upregulation of matrix metalloproteinase-2 and -9, indicating a potential fibrolytic effect. Immunomodulatory activities of CRT were also noted, as it increased cytokine expression in both macrophages and HpSCs. These effects were partially mediated through low-density lipoprotein receptor-related protein 1 (LRP1), as evidenced by altered cytokine expression upon co-treatment with a known LRP1 ligand receptor-associated protein (RAP). Overall, this study elucidates the complex role of extracellular CRT in liver injury and its potential impact on HpSC behavior and immune responses.
Collapse
Affiliation(s)
- Chien-Chih Chen
- Department of Psychiatry Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; School of Medicine Chang Gung University Taoyuan Taiwan
| | - Li-Wen Hsu
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - Kuang-Den Chen
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; Institute for Translational Research in Biomedicine Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - King-Wah Chiu
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; Division of Hepato-Gastroenterology Department of Internal Medicine Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - Chao-Pin Kung
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; Institute for Translational Research in Biomedicine Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - Shu-Rong Li
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - Shigeru Goto
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; Nobeoka Medical Check Center Fukuoka Institute of Occupational Health Nobeoka Japan
| | - Chao-Long Chen
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - Kuang-Tzu Huang
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; Institute for Translational Research in Biomedicine Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan.
| |
Collapse
|
48
|
Gui L, Chen K, Yan J, Chen P, Gao WQ, Ma B. Targeting the mevalonate pathway potentiates NUAK1 inhibition-induced immunogenic cell death and antitumor immunity. Cell Rep Med 2025; 6:101913. [PMID: 39824180 PMCID: PMC11866496 DOI: 10.1016/j.xcrm.2024.101913] [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: 06/07/2024] [Revised: 10/19/2024] [Accepted: 12/13/2024] [Indexed: 01/20/2025]
Abstract
The induction of immunogenic cell death (ICD) impedes tumor progression via both tumor cell-intrinsic and -extrinsic mechanisms, representing a robust therapeutic strategy. However, ICD-targeted therapy remains to be explored and optimized. Through kinome-wide CRISPR-Cas9 screen, NUAK family SNF1-like kinase 1 (NUAK1) is identified as a potential target. The ICD-provoking effect of NUAK1 inhibition depends on the production of reactive oxygen species (ROS), consequent to the downregulation of nuclear factor erythroid 2-related factor 2 (NRF2)-mediated antioxidant gene expression. Moreover, the mevalonate pathway/cholesterol biosynthesis, activated by spliced form of X-box binding protein 1 (XBP1s) downstream of ICD-induced endoplasmic reticulum (ER) stress, functions as a negative feedback mechanism. Targeting the mevalonate pathway with CRISPR knockout or the 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) inhibitor simvastatin amplifies NUAK1 inhibition-mediated ICD and antitumor activity, while cholesterol dampens ROS and ICD, and therefore also dampens tumor suppression. The combination of NUAK1 inhibitor and statin enhances the efficacy of anti-PD-1 therapy. Collectively, our study unveils the promise of blocking the mevalonate-cholesterol pathway in conjunction with ICD-targeted immunotherapy.
Collapse
Affiliation(s)
- Liming Gui
- Renji-Med-X Clinical Stem Cell Research Center, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Kaiwen Chen
- Renji-Med-X Clinical Stem Cell Research Center, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jingjing Yan
- Renji-Med-X Clinical Stem Cell Research Center, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ping Chen
- Renji-Med-X Clinical Stem Cell Research Center, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wei-Qiang Gao
- Renji-Med-X Clinical Stem Cell Research Center, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Bin Ma
- Renji-Med-X Clinical Stem Cell Research Center, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
49
|
Bekeschus S, Roessler K, Kepp O, Freund E. Gas Plasma Technology and Immunogenic Cell Death: Implications for Chordoma Treatment. Cancers (Basel) 2025; 17:681. [PMID: 40002275 PMCID: PMC11852646 DOI: 10.3390/cancers17040681] [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: 12/23/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer is the second-leading cause of death in developed societies. Specifically, cancers of the spine and brain come with significant therapeutic challenges. Chordomas are semi-malignant tumors that develop from embryonic residuals at the skull base (clival) or coccyx (sacral). Small tumor fragments can remain in the operation cavities during surgical resection, forming new tumor sites. This requires repeated surgeries or the application of proton-beam radiation and chemotherapy, which often do not lead to complete remission of the tumors. Hence, there is a need for novel therapeutic avenues that are not limited to killing visible tumors but can be applied after surgery to decrease chordoma recurrences. Reactive oxygen species (ROS) generated locally via novel medical gas plasma technologies are one potential approach to address this clinical problem. Previously, broad-spectrum free radicals generated by these cold physical plasmas operated at about body temperature were shown to oxidize cancer cells to the disadvantage of their growth and induce immunogenic cancer cell death (ICD), ultimately promoting anticancer immunity. This review outlines the clinical challenges of chordoma therapy, how medical gas plasma technology could serve as an adjuvant treatment modality, and potential immune-related mechanisms of action that could extend the longevity of gas plasma therapy beyond its acute local tissue effects.
Collapse
Affiliation(s)
- Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), 17489 Greifswald, Germany
- Department of Dermatology and Venerology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Karl Roessler
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75005 Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94800 Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, 94270 Kremlin Bicêtre, France
| | - Eric Freund
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
50
|
Zhou Y, Wei Y, Tian X, Wei X. Cancer vaccines: current status and future directions. J Hematol Oncol 2025; 18:18. [PMID: 39962549 PMCID: PMC11834487 DOI: 10.1186/s13045-025-01670-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
Cancer continues to be a major global health burden, with high morbidity and mortality. Building on the success of immune checkpoint inhibitors and adoptive cellular therapy, cancer vaccines have garnered significant interest, but their clinical success remains modest. Benefiting from advancements in technology, many meticulously designed cancer vaccines have shown promise, warranting further investigations to reach their full potential. Cancer vaccines hold unique benefits, particularly for patients resistant to other therapies, and they offer the ability to initiate broad and durable T cell responses. In this review, we highlight the antigen selection for cancer vaccines, introduce the immune responses induced by vaccines, and propose strategies to enhance vaccine immunogenicity. Furthermore, we summarize key features and notable clinical advances of various vaccine platforms. Lastly, we delve into the mechanisms of tumor resistance and explore the potential benefits of combining cancer vaccines with standard treatments and other immunomodulatory approaches to improve vaccine efficacy.
Collapse
Affiliation(s)
- Yingqiong Zhou
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|