1
|
Cai X, Liu Y, Luo G, Yu Z, Jiang C, Xu C. Ultrasound-assisted immunotherapy for malignant tumour. Front Immunol 2025; 16:1547594. [PMID: 40433381 PMCID: PMC12106521 DOI: 10.3389/fimmu.2025.1547594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Malignant tumour represents a significant global public health concern. The advent of immunotherapy has brought about a revolutionary shift in the landscape of tumour treatment, offering a ray of hope to patients across the globe. Immunotherapy strategies have demonstrated considerable promise in clinical trials. However, the immunosuppressive environment within the tumour microenvironment has constituted a significant obstacle to the advancement of immunotherapies. It is therefore imperative to develop more efficacious and personalised approaches. The utilisation of non-invasive ultrasound-assisted immunotherapy represents a promising strategy. Ultrasound has the capacity to induce an immune response and stimulate other drugs to achieve a specific response, thereby reducing the toxic side effects of treatment and enhancing the outcome of immunotherapy. This paper presents a systematic introduction to the various mechanisms related to ultrasound and reviews the recent advancements of ultrasound-assisted tumour immunotherapy, including ultrasonic ablation, combined application with contrast agents, and sonodynamic therapy.
Collapse
Affiliation(s)
- Xiaowen Cai
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yujie Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Guosheng Luo
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
| | - Zhanwang Yu
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
| | - Cheng Jiang
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
| | - Chuanshan Xu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
2
|
Chiu TY, Zho YJ, Ho YJ. Evaluating Immune Activation Feasibility in Pancreatic Ductal Adenocarcinoma via Oxygen Bubble-Induced Anti-Vascular Therapy. Pharmaceutics 2025; 17:645. [PMID: 40430936 PMCID: PMC12114744 DOI: 10.3390/pharmaceutics17050645] [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: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Anti-vascular therapy presents a potential strategy for activating anti-tumor immunity. Disrupted vascular debris provides effective antigens that activate dendritic cells, leading to subsequent immune responses. However, the resulting tumor hypoxia following vascular disruption may contribute to immune suppression, thereby hindering effective immune activation. Ultrasound-stimulated microbubble cavitation can locally disrupt tumor vessels through mechanical effects to achieve physical anti-vascular therapy. Therefore, this study designed oxygen-loaded nanobubbles (ONBs) to combine anti-vascular effects with local oxygen release under ultrasound stimulation. The feasibility of enhancing anti-tumor immune activation by alleviating tumor hypoxia was evaluated. Methods: A murine pancreatic subcutaneous solid tumor model was used to evaluate the efficacy of anti-vascular therapy-associated immunotherapy. Results: After ONB treatment, tumor perfusion was reduced to 52 ± 5%, which resulted in a subsequent 57 ± 11% necrosis and a 29 ± 4% reduction in hypoxia, demonstrating the anti-vascular effect and reoxygenation, respectively. However, subsequent immune responses exhibited no significant activation in intratumoral cytokine expression or splenic immune cell composition. Primary tumors exhibited a 15.7 ± 5.0% increase in necrosis following ONB treatment, but distant tumor growth was not significantly inhibited. Conclusions: These results highlighted a crucial issue regarding the complex correlations between vessel disruption, antigen production, oxygen delivery, hypoxia, and immunity when combining anti-vascular therapy with immunotherapy.
Collapse
Affiliation(s)
- Tzu-Yun Chiu
- Department of Biological Science and Technology, College of Engineering Bioscience, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
| | - Yi-Jia Zho
- Institute of Molecular Medicine and Bioengineering, College of Engineering Bioscience, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
| | - Yi-Ju Ho
- Department of Biological Science and Technology, College of Engineering Bioscience, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
| |
Collapse
|
3
|
Li X, Tan C, Fu X, Qiu J, Shen W, Xu Z, Wu X, Zhou Y, Li X, Sun L, Qin J. Disrupting Cdc42 activation-driven filopodia formation with low-intensity ultrasound and microbubbles: A novel strategy to block ovarian cancer metastasis. Colloids Surf B Biointerfaces 2025; 253:114724. [PMID: 40300280 DOI: 10.1016/j.colsurfb.2025.114724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/04/2025] [Accepted: 04/18/2025] [Indexed: 05/01/2025]
Abstract
Metastasis is a primary cause of mortality and treatment failure in ovarian cancer, with limited effective therapeutic strategies. Low-intensity ultrasound (LIUS) and microbubbles (MBs) has been demonstrated as an adjunctive technique capable of enhancing drug delivery and suppressing tumor metastasis. However, the underlying mechanisms remain incompletely understood. In this study, we aimed to investigate whether LIUS + MBs alone could suppress tumor metastasis and to explore its mechanism of action through disruption of the cytoskeletal remodeling in filopodia, an essential structure in the early stages of cancer cell dissemination. Based on cell-based experiments to determine the optimal parameters, our results showed LIUS + MBs significantly inhibited the migration and invasion of ovarian cancer cells. In vivo, LIUS + MBs treatment markedly suppressed the overall metastasis in the orthotopic ovarian cancer model, and in both the intraperitoneal and hematogenous metastatic models established by injecting pretreated cells. Morphologically, such treatment led to a notable reduction in the length and number of filopodia, while the number of lamellipodia remained unaffected. At the molecular level, LIUS + MBs disturbed filopodia formation and the metastatic potential of ovarian cancer cells by suppressing the activation of Cdc42, a key regulator of cytoskeletal dynamics. The inhibitory effect was reversed by the overexpression of Cdc42CA. Further proteomic and bioinformatics analysis implied that LIUS + MBs may reduce Cdc42 activity by upregulating the expression of GTPase-activating proteins (GAPs). Our research provides novel insight into the mechanism by which LIUS + MBs can inhibit tumor metastasis, highlighting its role in disturbing the Cdc42-mediated cytoskeletal remodelling of filopodia.
Collapse
Affiliation(s)
- Xiaoying Li
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou 310006, China; Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Chengwei Tan
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiuxiu Fu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Jian Qiu
- Department of Obstetrics and Gynaecology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Wanting Shen
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Zhikang Xu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaodong Wu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yiting Zhou
- Department of Orthopaedic Surgery and Department of Biochemistry of the Second Affiliated Hospital, Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Xiao Li
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, 310006, China; Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Hangzhou 310006, China.
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou 310006, China.
| | - Jiale Qin
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, 310006, China; Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Hangzhou 310006, China.
| |
Collapse
|
4
|
Hu C, Li H, Deng T, Liu Z, Yang L, Peng L, Jiang MY, Chen WZ. Abscopal effect of focused ultrasound combined immunotherapy in animal solid tumor model: a systematic reviews and meta-analysis. Front Immunol 2024; 15:1474343. [PMID: 39735534 PMCID: PMC11671366 DOI: 10.3389/fimmu.2024.1474343] [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: 08/01/2024] [Accepted: 11/11/2024] [Indexed: 12/31/2024] Open
Abstract
Introduction The abscopal effect, a systemic anti-tumor response triggered by localized treatment, has gained attention but remains poorly understood. This study evaluates the efficacy and consistency of focused ultrasound (FUS) combined with immunotherapy in inducing the abscopal effect. Methods A systematic review and meta-analysis were conducted on preclinical studies using solid tumor models. Data on tumor response, immune modulation, and survival outcomes were analyzed to assess the combination therapy's effectiveness. Results FUS combined with immunotherapy enhanced anti-tumor responses at local and distant sites, with evidence of immune activation and increased abscopal effect rates. However, heterogeneity across tumor models and protocols was observed. Discussion The findings provide a theoretical basis for FUS-immunotherapy combinations in cancer treatment, while emphasizing the need for standardized protocols and further research to elucidate underlying mechanisms. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023460710.
Collapse
Affiliation(s)
- Chao Hu
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing, China
- Department of Pulmonary and Critical Medicine, Xiangtan Central Hospital, Xiangtan, China
| | - Hui Li
- Department of Pulmonary and Critical Medicine, Xiangtan Central Hospital, Xiangtan, China
| | - Tao Deng
- Department of Pulmonary and Critical Medicine, Xiangtan Central Hospital, Xiangtan, China
| | - Zheng Liu
- Department of Pharmaceutical, Xiangtan Central Hospital, Xiangtan, China
| | - Li Yang
- Department of Pulmonary and Critical Medicine, Xiangtan Central Hospital, Xiangtan, China
| | - Li Peng
- Department of Oncology, Xiangtan Central Hospital, Xiangtan, China
| | - Ming Yan Jiang
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing, China
- Department of Pulmonary and Critical Medicine, Xiangtan Central Hospital, Xiangtan, China
| | - Wen Zhi Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Hu C, Fu Q, Gao FF, Zeng J, Xiao W, Li H, Peng L, Huang X, Yang L, Chen WZ, Jiang MY. Ultrasound-Guided High-Intensity Focused Ultrasound Combined With PD-1 Blockade in Patients With Liver Metastases From Lung Cancer: Protocol for a Single-Arm Phase 2 Trial. JMIR Res Protoc 2024; 13:e59152. [PMID: 39612480 PMCID: PMC11645512 DOI: 10.2196/59152] [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: 04/03/2024] [Revised: 10/01/2024] [Accepted: 10/26/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND While immunotherapy has revolutionized oncological management, its efficacy in lung cancer patients with liver metastases remains limited, potentially due to the unique immunosuppressive microenvironment of the liver. Local liver treatment has been shown to enhance the immunotherapy response, and high-intensity focused ultrasound (HIFU), a minimally invasive local treatment, has demonstrated promising results in combination with immunotherapy. However, clinical data regarding HIFU in lung cancer with liver metastases are limited. OBJECTIVE We designed the HILL (Ultrasound-Guided High-Intensity Focused Ultrasound Combined With PD-1 Blockade in Patients With Liver Metastases From Lung Cancer) study to investigate the effectiveness and safety of HIFU in combination with immunotherapy for lung cancer with liver metastases. METHODS The HILL study is a single-armed, single-center, phase 2 clinical trial that will enroll 30 patients with lung cancer and liver metastases. The treatment regimen involves administering HIFU to liver metastases 1 week before the first dose of a programmed cell death protein (PD)-1 blockade, which is then administered every 3 weeks. The primary aim is to determine the overall response rate based on immune-related response criteria. Secondary aims include safety, progression-free survival, overall response, overall survival, and quality of life. Exploratory studies will also be conducted using whole blood, plasma, archival cancer tissue, and tumor biopsies during progression or relapse to identify potential biomarkers. RESULTS The study was funded on March 14, 2022, and received ethical approval on April 27, 2022. Clinical trial registration was completed by June 10, 2022, with participant recruitment beginning on July 10, 2022. Data collection commenced on July 14, 2022, with the enrollment of the first patient. By April 2024, 6 participants had been recruited. The results are expected to be published in December 2026. CONCLUSIONS This study seeks to improve treatment outcomes for lung cancer patients with liver metastases by combining HIFU and PD-1 inhibition. The study also aims to identify potential biomarkers through exploratory research that can aid in selecting patients for optimized outcomes in the future. TRIAL REGISTRATION Chinese Clinical Trial Registry ChiCTR2200061076; https://www.chictr.org.cn/showproj.html?proj=170967. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/59152.
Collapse
Affiliation(s)
- Chao Hu
- Respiratory Department, Xiangtan Central Hospital, Xiangtan, China
| | - Qiang Fu
- Haifu Micro Noninvasive Center, Xiangtan Central Hospital, Xiangtan, China
| | - Fei Fei Gao
- Respiratory Department, Xiangtan Central Hospital, Xiangtan, China
| | - Jian Zeng
- Haifu Micro Noninvasive Center, Xiangtan Central Hospital, Xiangtan, China
| | - Wei Xiao
- Haifu Micro Noninvasive Center, Xiangtan Central Hospital, Xiangtan, China
| | - Hui Li
- Respiratory Department, Xiangtan Central Hospital, Xiangtan, China
| | - Li Peng
- Haifu Micro Noninvasive Center, Xiangtan Central Hospital, Xiangtan, China
| | - Xi Huang
- Respiratory Department, Xiangtan Central Hospital, Xiangtan, China
| | - Li Yang
- Respiratory Department, Xiangtan Central Hospital, Xiangtan, China
| | - Wen Zhi Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing, China
| | - Ming Yan Jiang
- Respiratory Department, Xiangtan Central Hospital, Xiangtan, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Liu D, Wang L, Guo Y. Advances in and prospects of immunotherapy for prostate cancer. Cancer Lett 2024; 601:217155. [PMID: 39127338 DOI: 10.1016/j.canlet.2024.217155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/07/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Immunotherapy has shown promising therapeutic effects in hematological malignancies and certain solid tumors and has emerged as a critical and highly potential treatment modality for cancer. However, prostate cancer falls under the category of immune-resistant cold tumors, for which immunotherapy exhibits limited efficacy in patients with solid tumors. Thus, it is important to gain a deeper understanding of the tumor microenvironment in prostate cancer to facilitate immune system activation and overcome immune suppression to advance immunotherapy for prostate cancer. In this review, we discuss the immunosuppressive microenvironment of prostate cancer, which is characterized by the presence of few tumor-infiltrating lymphocytes, abundant immunosuppressive cells, low immunogenicity, and a noninflammatory phenotype, which significantly influences the efficacy of immunotherapy for prostate cancer. Immunotherapy is mainly achieved by activating the host immune system and overcoming immunosuppression. In this regard, we summarize the therapeutic advances in immune checkpoint blockade, immunogenic cell death, reversal of the immunosuppressive tumor microenvironment, tumor vaccines, immune adjuvants, chimeric antigen receptor T-cell therapy, and overcoming penetration barriers in prostate cancer, with the aim of providing novel research insights and approaches to enhance the effectiveness of immunotherapy for prostate cancer.
Collapse
Affiliation(s)
- Deng Liu
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China; Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Luofu Wang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
7
|
Guo D, Sheng K, Zhang Q, Li P, Sun H, Wang Y, Lyu X, Jia Y, Wang C, Wu J, Zhang X, Wang D, Sun Y, Huang S, Yu J, Zhang J. Single-cell transcriptomic analysis reveals the landscape of epithelial-mesenchymal transition molecular heterogeneity in esophageal squamous cell carcinoma. Cancer Lett 2024; 587:216723. [PMID: 38342234 DOI: 10.1016/j.canlet.2024.216723] [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: 11/11/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/13/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a prevalent and highly lethal malignant disease. The epithelial-mesenchymal transition (EMT) is crucial in promoting ESCC development. However, the molecular heterogeneity of ESCC and the potential inhibitory strategies targeting EMT remain poorly understood. In this study, we analyzed high-resolution single-cell transcriptome data encompassing 209,231 ESCC cells from 39 tumor samples and 16 adjacent samples obtained from 44 individuals. We identified distinct cell populations exhibiting heterogeneous EMT characteristics and identified 87 EMT-associated molecules. The expression profiles of these EMT-associated molecules showed heterogeneity across different stages of ESCC progression. Moreover, we observed that EMT primarily occurred in early-stage tumors, before lymph node metastasis, and significantly promoted the rapid deterioration of ESCC. Notably, we identified SERPINH1 as a potential novel marker for ESCC EMT. By classifying ESCC patients based on EMT gene sets, we found that those with high EMT exhibited poorer prognosis. Furthermore, we predicted and experimentally validated drugs targeting ESCC EMT, including dactolisib, docetaxel, and nutlin, which demonstrated efficacy in inhibiting EMT and metastasis in ESCC. Through the integration of scRNA-seq, RNA-seq, and TCGA data with experimental validation, our comprehensive analysis elucidated the landscape of EMT during the entire course of ESCC development and metastasis. These findings provide valuable insights and a reference for refining ESCC clinical treatment strategies.
Collapse
Affiliation(s)
- Dianhao Guo
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Kaiwen Sheng
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Qi Zhang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Pin Li
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Haoqiang Sun
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Yongjie Wang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Xinxing Lyu
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Yang Jia
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250117, China.
| | - Caifan Wang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Jing Wu
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Xiaohang Zhang
- Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau.
| | - Dandan Wang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Yawen Sun
- Department of Clinical Epidemiology and Biostatistics, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Shuhong Huang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| | - Jingze Zhang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
8
|
Chen L, Hou P, Zou YL, Wang Y, Zhou LL, Hu L, Hu Y, Zhang QY, Huang LP, Lin L. B7-H1 agonists suppress the PI3K/AKT/mtor pathway by degrading p110γ and independently induce cell death. Cancer Lett 2024; 584:216615. [PMID: 38199586 DOI: 10.1016/j.canlet.2024.216615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/03/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024]
Abstract
The biological role of B7-H1 intrinsic signal is reportedly diverse and controversial, its signal pathway remains unclear. Although B7-H1 blocking antibodies were found to have agonist capacity, their binding features and agonist mechanisms need further investigation. Here, by constructing cell strains with full-length or truncated B7-H1, we found that B7-H1 functioned as a receptor to transmit cell death signal from PD-1 protein or anti-B7-H1s through its cytoplasmic domain. Specific binding to the IgV-like domain of B7-H1 was required for the downstream signal. Upon agonists interaction, B7-H1 regulated the degradation of phosphoinositide 3-kinases (PI3Ks) subunit p110γ, subsequently inhibited the PI3K/AKT/mTOR pathway, and significantly increased autophagy. Moreover, B7-H1 agonists also suppressed ubiquitylation in B7-H1+cells by reducing ubiquitin-activating enzyme (E1), eventually leading to cell death. Finally, we validated the receptor role of B7-H1 in multiple tumor cells and demonstrated that B7-H1 agonists could suppress tumor progression independent of T cells in vivo. Our findings revealed that B7-H1 agonists functions as a PI3K inhibitor and may offer new strategies for PI3K targeting therapy.
Collapse
Affiliation(s)
- Ling Chen
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, 350102, China.
| | - Ping Hou
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, 350102, China
| | - Yu-Lian Zou
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, 350102, China
| | - Yang Wang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, 350102, China
| | - Lin-Lin Zhou
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, 350102, China
| | - Li Hu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, 350102, China
| | - Yan Hu
- Public Technology Service Center, Fujian Medical University, Fuzhou, Fujian, 350102, China
| | - Qiu-Yu Zhang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, 350102, China; Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350102, China
| | - Li-Ping Huang
- Department of Obstetrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Lin Lin
- Department of Obstetrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
| |
Collapse
|
9
|
Wu Y, Li J, Shu L, Tian Z, Wu S, Wu Z. Ultrasound combined with microbubble mediated immunotherapy for tumor microenvironment. Front Pharmacol 2024; 15:1304502. [PMID: 38487163 PMCID: PMC10937735 DOI: 10.3389/fphar.2024.1304502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/11/2024] [Indexed: 03/17/2024] Open
Abstract
The tumor microenvironment (TME) plays an important role in dynamically regulating the progress of cancer and influencing the therapeutic results. Targeting the tumor microenvironment is a promising cancer treatment method in recent years. The importance of tumor immune microenvironment regulation by ultrasound combined with microbubbles is now widely recognized. Ultrasound and microbubbles work together to induce antigen release of tumor cell through mechanical or thermal effects, promoting antigen presentation and T cells' recognition and killing of tumor cells, and improve tumor immunosuppression microenvironment, which will be a breakthrough in improving traditional treatment problems such as immune checkpoint blocking (ICB) and himeric antigen receptor (CAR)-T cell therapy. In order to improve the therapeutic effect and immune regulation of TME targeted tumor therapy, it is necessary to develop and optimize the application system of microbubble ultrasound for organs or diseases. Therefore, the combination of ultrasound and microbubbles in the field of TME will continue to focus on developing more effective strategies to regulate the immunosuppression mechanisms, so as to activate anti-tumor immunity and/or improve the efficacy of immune-targeted drugs, At present, the potential value of ultrasound combined with microbubbles in TME targeted therapy tumor microenvironment targeted therapy has great potential, which has been confirmed in the experimental research and application of breast cancer, colon cancer, pancreatic cancer and prostate cancer, which provides a new alternative idea for clinical tumor treatment. This article reviews the research progress of ultrasound combined with microbubbles in the treatment of tumors and their application in the tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | - Zuohui Wu
- Department of Ultrasound, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
10
|
Xie X, Zhang J, Wang Y, Shi W, Tang R, Tang Q, Sun S, Wu R, Xu S, Wang M, Liang X, Cui L. Nanomaterials augmented bioeffects of ultrasound in cancer immunotherapy. Mater Today Bio 2024; 24:100926. [PMID: 38179429 PMCID: PMC10765306 DOI: 10.1016/j.mtbio.2023.100926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/30/2023] [Accepted: 12/18/2023] [Indexed: 01/06/2024] Open
Abstract
Immunotherapy as a milestone in cancer treatment has made great strides in the past decade, but it is still limited by low immune response rates and immune-related adverse events. Utilizing bioeffects of ultrasound to enhance tumor immunotherapy has attracted more and more attention, including sonothermal, sonomechanical, sonodynamic and sonopiezoelectric immunotherapy. Moreover, the emergence of nanomaterials has further improved the efficacy of ultrasound mediated immunotherapy. However, most of the summaries in this field are about a single aspect of the biological effects of ultrasound, which is not comprehensive and complete currently. This review proposes the recent progress of nanomaterials augmented bioeffects of ultrasound in cancer immunotherapy. The concept of immunotherapy and the application of bioeffects of ultrasound in cancer immunotherapy are initially introduced. Then, according to different bioeffects of ultrasound, the representative paradigms of nanomaterial augmented sono-immunotherapy are described, and their mechanisms are discussed. Finally, the challenges and application prospects of nanomaterial augmented ultrasound mediated cancer immunotherapy are discussed in depth, hoping to pave the way for cancer immunotherapy and promote the clinical translation of ultrasound mediated cancer immunotherapy through the reasonable combination of nanomaterials augmented ultrasonic bioeffects.
Collapse
Affiliation(s)
- Xinxin Xie
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Jinxia Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Yuan Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Wanrui Shi
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Rui Tang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Qingshuang Tang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Suhui Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Ruiqi Wu
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Shuyu Xu
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Mengxin Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Ligang Cui
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| |
Collapse
|
11
|
Osada T, Jiang X, Zhao Y, Chen M, Kreager BC, Wu H, Kim H, Ren J, Snyder J, Zhong P, Morse MA, Lyerly HK. The use of histotripsy as intratumoral immunotherapy beyond tissue ablation-the rationale for exploring the immune effects of histotripsy. Int J Hyperthermia 2023; 40:2263672. [PMID: 37806666 DOI: 10.1080/02656736.2023.2263672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023] Open
Abstract
Mechanical high-intensity focused ultrasound (M-HIFU), which includes histotripsy, is a non-ionizing, non-thermal ablation technology that can be delivered by noninvasive methods. Because acoustic cavitation is the primary mechanism of tissue disruption, histotripsy is distinct from the conventional HIFU techniques resulting in hyperthermia and thermal injury. Phase I human trials have shown the initial safety and efficacy of histotripsy in treating patients with malignant liver tumors. In addition to tissue ablation, a promising benefit of M-HIFU has been stimulating a local and systemic antitumor immune response in preclinical models and potentially in the Phase I trial. Preclinical studies combining systemic immune therapies appear promising, but clinical studies of combinations have been complicated by systemic toxicities. Consequently, combining M-HIFU with systemic immunotherapy has been demonstrated in preclinical models and may be testing in future clinical studies. An additional alternative is to combine intratumoral M-HIFU and immunotherapy using microcatheter-placed devices to deliver both M-HIFU and immunotherapy intratumorally. The promise of M-HIFU as a component of anti-cancer therapy is promising, but as forms of HIFU are tested in preclinical and clinical studies, investigators should report not only the parameters of the energy delivered but also details of the preclinical models to enable analysis of the immune responses. Ultimately, as clinical trials continue, clinical responses and immune analysis of patients undergoing M-HIFU including forms of histotripsy will provide opportunities to optimize clinical responses and to optimize application and scheduling of M-HIFU in the context of the multi-modality care of the cancer patient.
Collapse
Affiliation(s)
- Takuya Osada
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Xiaoning Jiang
- Department of Mechanical and Aerospace Engineering, College of Engineering, NC State University, Raleigh, NC, USA
| | | | - Mengyue Chen
- Department of Mechanical and Aerospace Engineering, College of Engineering, NC State University, Raleigh, NC, USA
| | - Benjamin C Kreager
- Department of Mechanical and Aerospace Engineering, College of Engineering, NC State University, Raleigh, NC, USA
| | - Huaiyu Wu
- Department of Mechanical and Aerospace Engineering, College of Engineering, NC State University, Raleigh, NC, USA
| | - Howuk Kim
- Department of Mechanical Engineering, School of Engineering, Inha University, Incheon, Republic of South Korea
| | - Jun Ren
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Joshua Snyder
- Department of Surgery and Cell Biology, Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Pei Zhong
- Thomas Lord Department of Mechanical Engineering and Material Science, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Michael A Morse
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, NC, USA
| | - H Kim Lyerly
- Department of Surgery, Pathology, and Integrative Immunobiology, Duke University School of Medicine, Duke University, Durham, NC, USA
| |
Collapse
|
12
|
Wang C, Wu L, Yuan H, Yu H, Xu J, Chen S, Yan S, Wang X. A powerful antitumor "trident": the combination of radio-, immuno- and anti-angiogenesis therapy based on mesoporous silica single coated gold nanoparticles. J Mater Chem B 2023; 11:879-889. [PMID: 36594928 DOI: 10.1039/d2tb02046g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although immunotherapy in combination with anti-angiogenesis therapy has made a breakthrough in the first-line treatment of cancer, considering the low responder rate and the adverse events, it is vital to propose a new combination modality. In this study, we report single encapsulated mesoporous silica coated gold nanoparticles that synergize sensitizing radiotherapy with the current combination therapy. Distinguished from simply combining two treatments, the nanoparticle-mediated "trident" therapy resolved the problem of matching the dose between radiation and drug, which determines the outcome since drug demand rises with immunosuppression from increased sensitivity to radiotherapy. The nanomedicine produced energy depositions when radiation was introduced, and released the loaded toripalimab and bevacizumab, exhibiting significant anti-tumor properties. In vitro tumor cell viability results indicated the highest inhibition by the "trident" therapy and in vivo animal models also revealed the earliest decrease in tumor tissue volume. As a result, the "trident" therapy is expected to further improve the anti-tumor benefits of the combination of immunotherapy and anti-angiogenesis therapy and provides a versatile perspective on cancer treatment.
Collapse
Affiliation(s)
- Cheng Wang
- College of Materials Science & Engineering, Zhejiang University of Technology, China.
| | - Lingyun Wu
- The First Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Huili Yuan
- College of Materials Science & Engineering, Zhejiang University of Technology, China.
| | - Hao Yu
- The First Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Jiaqi Xu
- The First Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Si Chen
- College of Materials Science & Engineering, Zhejiang University of Technology, China.
| | - Senxiang Yan
- The First Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Xu Wang
- College of Materials Science & Engineering, Zhejiang University of Technology, China.
| |
Collapse
|