1
|
Shi X, Askari Rizvi SF, Yang Y, Liu G. Emerging nanomedicines for macrophage-mediated cancer therapy. Biomaterials 2025; 316:123028. [PMID: 39693782 DOI: 10.1016/j.biomaterials.2024.123028] [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: 10/18/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Tumor-associated macrophages (TAMs) contribute to tumor progression by promoting angiogenesis, remodeling the tumor extracellular matrix, inducing tumor invasion and metastasis, as well as immune evasion. Due to the high plasticity of TAMs, they can polarize into different phenotypes with distinct functions, which are primarily categorized as the pro-inflammatory, anti-tumor M1 type, and the anti-inflammatory, pro-tumor M2 type. Notably, anti-tumor macrophages not only directly phagocytize tumor cells, but also present tumor-specific antigens and activate adaptive immunity. Therefore, targeted regulation of TAMs to unleash their potential anti-tumor capabilities is crucial for improving the efficacy of cancer immunotherapy. Nanomedicine serves as a promising vehicle and can inherently interact with TAMs, hence, emerging as a new paradigm in cancer immunotherapy. Due to their controllable structures and properties, nanomedicines offer a plethora of advantages over conventional drugs, thus enhancing the balance between efficacy and toxicity. In this review, we provide an overview of the hallmarks of TAMs and discuss nanomedicines for targeting TAMs with a focus on inhibiting recruitment, depleting and reprogramming TAMs, enhancing phagocytosis, engineering macrophages, as well as targeting TAMs for tumor imaging. We also discuss the challenges and clinical potentials of nanomedicines for targeting TAMs, aiming to advance the exploitation of nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Xueying Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Syed Faheem Askari Rizvi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China; Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Punjab, Pakistan
| | - Yinxian Yang
- School of Pharmaceutical Sciences, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
2
|
Liu Q, Yu M, Lin Z, Wu L, Xia P, Zhu M, Huang B, Wu W, Zhang R, Li K, Zhu L, Wang Q. COL1A1-positive endothelial cells promote gastric cancer progression via the ANGPTL4-SDC4 axis driven by Endothelial-to-Mesenchymal Transition. Cancer Lett 2025:217731. [PMID: 40254092 DOI: 10.1016/j.canlet.2025.217731] [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: 10/31/2024] [Revised: 03/19/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Gastric cancer (GC) is an aggressive and heterogeneous disease with poor survival outcomes. The progression of GC involves complex, multi-step processes. Endothelial cells (ECs) play a crucial role in tumor angiogenesis, proliferation, invasion, and metastasis, particularly through the process of endothelial-to-mesenchymal transition (EndoMT). However, the specific role and mechanisms of EndoMT in gastric cancer remain unclear. Based on 6 GC single-cell RNA-sequencing (scRNA-seq) cohorts (samples = 97), we established a EndoMT-related gene signature, termed EdMTS. Leveraging this gene signature, ssGSEA was applied to calculate sample scores across multiple bulk RNA-seq datasets, which include information on immunotherapy, metastasis, GC progression, and survival. Moreover, we applied the Monocle2 method to calculate cell pseudotime and used CellChat to analyze interactions between malignant and EC cells. We verified the molecular mechanism by multiple immunofluorescence and cell function experiments. Findings In this study, we established a single-cell atlas of ECs in GC and identified a subpopulation of COL1A1+ ECs that play a critical role in tumor progression and metastasis. These COL1A1+ ECs were significantly associated with worse clinical outcomes in GC patients. Further analysis revealed that COL1A1+ ECs originated from lymphatic ECs and underwent EndoMT through the upregulation of CEBPB, driving tumor invasiveness. Moreover, COL1A1+ ECs interacted with malignant cells via ANGPTL4-SDC4 axis, enhancing invasion and migration. These findings provide a deeper understanding of the role of COL1A1+ ECs in GC progression and highlight potential therapeutic targets for disrupting the EndoMT process in these cells to provide a benefit for GC patients.
Collapse
Affiliation(s)
- Quanzhong Liu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002 Nanjing, China
| | - Miao Yu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Zihan Lin
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Lingxiang Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002 Nanjing, China
| | - Peng Xia
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Mengyan Zhu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002 Nanjing, China
| | - Bin Huang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002 Nanjing, China
| | - Wei Wu
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Ruohan Zhang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002 Nanjing, China
| | - Kening Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002 Nanjing, China
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002 Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
3
|
Wang W, Zhai Y, Yang X, Ye L, Lu G, Shi X, Zhai G. Effective design of therapeutic nanovaccines based on tumor neoantigens. J Control Release 2025; 380:17-35. [PMID: 39892648 DOI: 10.1016/j.jconrel.2025.01.078] [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: 09/05/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Neoantigen vaccines are among the most potent immunotherapies for personalized cancer treatment. Therapeutic vaccines containing tumor-specific neoantigens that elicit specific T cell responses offer the potential for long-term clinical benefits to cancer patients. Unlike immune-checkpoint inhibitors (ICIs), which rely on pre-existing specific T cell responses, personalized neoantigen vaccines not only promote existing specific T cell responses but importantly stimulate the generation of neoantigen-specific T cells, leading to the establishment of a persistent specific memory T cell pool. The review discusses the current state of clinical research on neoantigen nanovaccines, focusing on the application of vectors, adjuvants, and combinational strategies to address a range of challenges and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Weilin Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States of America
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
4
|
Cousin S, Guégan JP, Palmieri LJ, Metges JP, Pernot S, Bellera CA, Assenat E, Korakis I, Cassier PA, Hollebecque A, Cantarel C, Kind M, Soubeyran I, Sokol H, Vanhersecke L, Bessede A, Italiano A. Regorafenib plus avelumab in advanced gastroenteropancreatic neuroendocrine neoplasms: a phase 2 trial and correlative analysis. NATURE CANCER 2025:10.1038/s43018-025-00916-3. [PMID: 40204996 DOI: 10.1038/s43018-025-00916-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 01/17/2025] [Indexed: 04/11/2025]
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are heterogeneous tumors with limited treatment options. This phase 2 Bayesian study evaluated the combination of regorafenib, a multikinase inhibitor, and avelumab, a programmed death 1 (PD1) ligand 1 inhibitor, in advanced grade 2-grade 3 well-differentiated GEP neuroendocrine tumors or grade 3 GEP neuroendocrine carcinomas after progression on prior therapies. A total of 47 participants were enrolled and 42 were evaluable for efficacy. Participants received regorafenib (160 mg per day) and avelumab (10 mg kg-1 biweekly) in 28-day cycles. The primary endpoint, 6-month objective response rate per the response evaluation criteria in solid tumors version 1.1, was 18% (95% confidence interval (CI): 8-31%), with a median progression-free survival of 5.5 months (95% CI: 3.6-8). Durable responses were noted (16.6 months; 95% CI: 3.7-no response). Treatment-related adverse events were manageable, with fatigue, diarrhea and palmar-plantar erythrodysesthesia being most common. Exploratory biomarker analysis identified PD1 and indoleamine 2,3-dioxygenase 1 expression and activity as potential resistance markers. These findings highlight the clinical potential of regorafenib and avelumab in GEP-NENs, emphasizing the need for predictive biomarkers and validation in future randomized trials. Clinical Trial registration: NCT03475953 .
Collapse
Affiliation(s)
- Sophie Cousin
- Department of Medicine, Institut Bergonié, Bordeaux, France
| | | | - Lola Jade Palmieri
- Department of Medicine, Institut Bergonié, Bordeaux, France
- Explicyte, Bordeaux, France
| | | | - Simon Pernot
- Department of Medicine, Institut Bergonié, Bordeaux, France
| | - Carine A Bellera
- INSERM, Bordeaux Population Health Research Center, Epicene team, University of Bordeaux, UMR 1219, Bordeaux, France
- INSERM CIC1401, Clinical and Epidemiological Research Unit, Bordeaux, France
| | - Eric Assenat
- Department of Medicine, Institut Cancérologie Montpellier, Montpellier, France
| | | | | | | | - Coralie Cantarel
- INSERM CIC1401, Clinical and Epidemiological Research Unit, Bordeaux, France
| | - Michèle Kind
- Department of Radiology, Institut Bergonié, Bordeaux, France
| | | | - Harry Sokol
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Gastroentérologie, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | | | - Antoine Italiano
- Department of Medicine, Institut Bergonié, Bordeaux, France.
- DITEP, Gustave Roussy, Villejuif, France.
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Gastroentérologie, Paris, France.
- Faculty of Medicine, University of Bordeaux, Bordeaux, France.
| |
Collapse
|
5
|
Zhong X, Fei Y, Zhao H, Chen J, Gao M, Huang Y, Fei W. Mechanistic studies and therapeutic potential of angiopoietin in head and neck tumor angiogenesis. Front Oncol 2025; 15:1529225. [PMID: 40260291 PMCID: PMC12010120 DOI: 10.3389/fonc.2025.1529225] [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: 11/19/2024] [Accepted: 02/21/2025] [Indexed: 04/23/2025] Open
Abstract
Head and neck tumors represent a prevalent category of oral and maxillofacial malignancies, posing significant therapeutic and prognostic challenges due to their complex anatomical structure, tumor heterogeneity, and resistance to conventional therapies. Recent studies have highlighted the strong association between tumor progression and neoangiogenesis, with the angiopoietin (ANG) family playing a central role in this process. Comprising ANG1, ANG2, ANG3, and ANG4, these factors regulate multiple signaling pathways that promote cellular growth, differentiation, and proliferation, thereby driving angiogenesis and accelerating tumor growth and metastasis. Therefore, a comprehensive investigation of the ANG family's role in head and neck tumors may offer critical insights into tumorigenesis mechanisms and unveil novel therapeutic targets. Such research has the potential to improve treatment outcomes and enhance the quality of life for patients.
Collapse
Affiliation(s)
- Xiaojuan Zhong
- School of Medicine, University of Electronic Science and Technology, Chengdu, Sichuan, China
| | - Yujie Fei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Haihui Zhao
- School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Jiao Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Mingyu Gao
- Yibin Second People’s Hospital, Yibin, Sichuan, China
| | - Yi Huang
- Department of Maxillofacial Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Fei
- Department of Maxillofacial Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Oral and Maxillofacial Surgery, Wenjiang Hospital, Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Ultimo A, Jain A, Gomez-Gonzalez E, Alex TS, Moreno-Borrallo A, Jana S, Ghosh S, Ruiz-Hernandez E. Nanotherapeutic Formulations for the Delivery of Cancer Antiangiogenics. Mol Pharm 2025. [PMID: 40184281 DOI: 10.1021/acs.molpharmaceut.4c00822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2025]
Abstract
Antiangiogenic medications for cancer treatment have generally failed in showing substantial benefits in terms of prolonging life on their own; their effects are noticeable only when combined with chemotherapy. Moreover, treatments based on prolonged antiangiogenics administration have demonstrated to be ineffective in stopping tumor progression. In this scenario, nanotherapeutics can address certain issues linked to existing antiangiogenic treatments. More specifically, they can provide the ability to target the tumor's blood vessels to enhance drug accumulation and manage release, ultimately decreasing undesired side effects. Additionally, they enable the administration of multiple angiogenesis inhibitors at the same time as chemotherapy. Key reports in this field include the design of polymeric nanoparticles, inorganic nanoparticles, vesicles, and hydrogels for loading antiangiogenic substances like endostatin and interleukin-12. Furthermore, nanoformulations have been proposed to efficiently control relevant pro-angiogenic pathways such as VEGF, Tie2/Angiopoietin-1, HIF-1α/HIF-2α, and TGF-β, providing powerful approaches to block tumor growth and metastasis. In this article, we outline a selection of nanoformulations for antiangiogenic treatments for cancer that have been developed in the past ten years.
Collapse
Affiliation(s)
- Amelia Ultimo
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Ayushi Jain
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Elisabet Gomez-Gonzalez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Thomson Santosh Alex
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Almudena Moreno-Borrallo
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Sukanya Jana
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Shubhrima Ghosh
- Trinity Translational Medicine Institute, Trinity College Dublin, the University of Dublin, St. James's Hospital, Dublin 8 D08 NHY1, Ireland
- School of Biological, Health and Sports Sciences, Technological University Dublin, Grangegorman Lower, Dublin 7 D07 ADY7, Ireland
| | - Eduardo Ruiz-Hernandez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| |
Collapse
|
7
|
Hu WT, Li M, Ma PJ, Yang D, Liu XD, Wang Y. A silence catalyst: CCL5-mediated intercellular communication in cancer. Arch Toxicol 2025:10.1007/s00204-025-04036-w. [PMID: 40167774 DOI: 10.1007/s00204-025-04036-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Chemokine CCL5 (RANTES), as a key mediator of intercellular communication in cancers, and its role in cancer development, metastasis and immune escape has received increasing attention. CCL5 and its receptors are important components of the tumor microenvironment and play a tumor promoting role in different ways by triggering signaling pathways through binding to the primary receptor CCR5. CCL5 was viewed as indispensable "gate keepers" of immunity and inflammation, it remains unclear of CCL5-mediated intercellular communication. Therefore, in this review, we summarize the latest information on the origin, structure, and characterization of CCL5 and role of CCL5 in the tumor microenvironment. It includes CCL5-mediated intercellular communication through exosomes, microvesicles and others in breast, lung, and ovarian cancers. CCL5 has a multifaceted role in cancer and has potential applications as a biomarker for cancer diagnosis and prognosis, which provides theoretical bases and therapeutic targets for the development of new cancer therapeutic strategies.
Collapse
Affiliation(s)
- Wei-Ting Hu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Ming Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Pei-Jun Ma
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Ding Yang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Xiao-Dong Liu
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
8
|
Hong R, Yu P, Zhang X, Su P, Liang H, Dong D, Wang X, Wang K. The role of cancer-associated fibroblasts in the tumour microenvironment of urinary system. Clin Transl Med 2025; 15:e70299. [PMID: 40195290 PMCID: PMC11975626 DOI: 10.1002/ctm2.70299] [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: 11/19/2024] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/09/2025] Open
Abstract
Urological tumours are a type of neoplasms that significantly jeopardise human life and wellbeing. Cancer-associated fibroblasts (CAFs), serving as the primary component of the stromal cellular milieu, form a diverse cellular cohort that exerts substantial influence on tumourigenesis and tumour progression. In this review, we summarised the literatures regarding the functions of CAFs in the urinary tumour microenvironment (TME). We primarily examined the multifaceted activities of CAFs in the TME of urological system tumours, including inhibiting tumour immunity, remodelling the extracellular matrix, promoting tumour growth, metastasis, drug resistance and their clinical applications. We also discussed potential future directions for leveraging artificial intelligence in CAFs research. KEY POINTS: The interaction of CAFs with various cell secretory factors in the TME of urological tumors. The application of CAFs in diagnosis, treatment and prognosis of urological tumors.
Collapse
Affiliation(s)
- Ri Hong
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Puguang Yu
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Xiaoli Zhang
- Department of Critical Care MedicineShengjing Hospital of China Medical UniversityShenyangChina
| | - Peng Su
- Medical Research CenterShengjing Hospital of China Medical UniversityShenyangChina
| | - Hongyuan Liang
- Department of RadiologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Dan Dong
- College of Basic Medical ScienceChina Medical UniversityShenyangChina
| | - Xuesong Wang
- Department of UrologyPeople's Hospital of China Medical UniversityShenyangChina
- Department of UrologyPeople's Hospital of Liaoning ProvinceShenyangChina
| | - Kefeng Wang
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
9
|
Chen L, Li L, Zhao H, Li H, Li J, Li C, Zhou Y, Yang L, Liang J, Zhang H, Li J, Xu P, Yuan C, Liu Z, Huang M, Jiang L. Integration of EMAP-II-targeted anti-angiogenesis and photodynamic therapy using zinc phthalocyanine nanosystem for enhanced cancer treatment. Colloids Surf B Biointerfaces 2025; 248:114493. [PMID: 39778222 DOI: 10.1016/j.colsurfb.2024.114493] [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: 10/03/2024] [Revised: 12/17/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Angiogenesis provides essential nutrients and oxygen to tumors during tumorigenesis, facilitating invasion and metastasis. Consequently, inhibiting tumor angiogenesis is an established strategy in anti-cancer therapy. In this study, we engineered a dual-function nanosystem with both antiangiogenic and photodynamic properties. We transformed the hydrophobic photosensitizer zinc phthalocyanine (PS) into a hydrophilic form via protein renaturation, resulting in a novel photosensitizer: Monocyte-Activating Polypeptide-II (EMAP-II:PS@NPs). Characterization through dynamic light scattering (DLS) and UV-vis spectroscopy showed that these nanoparticles exhibited uniform size and stability, and enhanced solubility. We further demonstrated that EMAP-II:PS@NPs effectively target tumor vascular endothelia causing intracellular photodynamic cytotoxicity. Notably, EMAP-II:PS@NPs achieved effective ablation of solid tumors at significantly reduced dosages of drugs compared to conventional therapies, due to their potent apoptotic effects on light-exposed cells. This study highlights the potential of combining anti-angiogenic activity with phototherapy, paving the way for innovative cancer treatment strategies.
Collapse
Affiliation(s)
- Liyun Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Linlin Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Hailong Zhao
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Hao Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Jiahui Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Chao Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Yang Zhou
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Luxuan Yang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Jun Liang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Honglian Zhang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Juan Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350116, China; The National & Local Joint Engineering Research Center on Biopharmaceutical and Photodynamic Therapy Technologies, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350116, China; The National & Local Joint Engineering Research Center on Biopharmaceutical and Photodynamic Therapy Technologies, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Zhenhua Liu
- Department of Oncology, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, Fujian 350001, China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China; The National & Local Joint Engineering Research Center on Biopharmaceutical and Photodynamic Therapy Technologies, Fuzhou University, Fuzhou, Fujian 350116, China.
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China; The National & Local Joint Engineering Research Center on Biopharmaceutical and Photodynamic Therapy Technologies, Fuzhou University, Fuzhou, Fujian 350116, China.
| |
Collapse
|
10
|
Di Spirito A, Balkhi S, Vivona V, Mortara L. Key immune cells and their crosstalk in the tumor microenvironment of bladder cancer: insights for innovative therapies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002304. [PMID: 40177538 PMCID: PMC11964778 DOI: 10.37349/etat.2025.1002304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Bladder cancer (BC) is a heterogeneous disease associated with high mortality if not diagnosed early. BC is classified into non-muscle-invasive BC (NMIBC) and muscle-invasive BC (MIBC), with MIBC linked to poor systemic therapy response and high recurrence rates. Current treatments include transurethral resection with Bacillus Calmette-Guérin (BCG) therapy for NMIBC and radical cystectomy with chemotherapy and/or immunotherapy for MIBC. The tumor microenvironment (TME) plays a critical role in cancer progression, metastasis, and therapeutic efficacy. A comprehensive understanding of the TME's complex interactions holds substantial translational significance for developing innovative treatments. The TME can contribute to therapeutic resistance, particularly in immune checkpoint inhibitor (ICI) therapies, where resistance arises from tumor-intrinsic changes or extrinsic TME factors. Recent advancements in immunotherapy highlight the importance of translational research to address these challenges. Strategies to overcome resistance focus on remodeling the TME to transform immunologically "cold" tumors, which lack immune cell infiltration, into "hot" tumors that respond better to immunotherapy. These strategies involve disrupting cancer-microenvironment interactions, inhibiting angiogenesis, and modulating immune components to enhance anti-tumor responses. Key mechanisms include cytokine involvement [e.g., interleukin-6 (IL-6)], phenotypic alterations in macrophages and natural killer (NK) cells, and the plasticity of cancer-associated fibroblasts (CAFs). Identifying potential therapeutic targets within the TME can improve outcomes for MIBC patients. This review emphasizes the TME's complexity and its impact on guiding novel therapeutic approaches, offering hope for better survival in MIBC.
Collapse
Affiliation(s)
- Anna Di Spirito
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Veronica Vivona
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
11
|
Zhang L, Zhao J, Su C, Wu J, Jiang L, Chi H, Wang Q. Organoid models of ovarian cancer: resolving immune mechanisms of metabolic reprogramming and drug resistance. Front Immunol 2025; 16:1573686. [PMID: 40191206 PMCID: PMC11968360 DOI: 10.3389/fimmu.2025.1573686] [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: 02/09/2025] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Metabolic reprogramming is a hallmark of ovarian cancer, enabling tumor progression, immune evasion and drug resistance. The tumor microenvironment (TME) further shapes metabolic adaptations, enabling cancer cells to withstand hypoxia and nutrient deprivation. While organoid models provide a physiologically relevant platform for studying these processes, they still lack immune and vascular components, limiting their ability to fully recapitulate tumor metabolism and drug responses. In this study, we investigated the key metabolic mechanisms involved in ovarian cancer progression, focusing on glycolysis, lipid metabolism and amino acid metabolism. We integrated metabolomic analyses and drug sensitivity assays to explore metabolic-TME interactions using patient-derived, adult stem cell-derived and iPSC-derived organ tissues. Among these, we found that glycolysis, lipid metabolism and amino acid metabolism play a central role in tumor progression and chemotherapy resistance. We identified methylglyoxal (MGO)-mediated BRCA2 dysfunction as a driver of immune escape, a role for sphingolipid signaling in tumor proliferation and a role for kynurenine metabolism in CD8+ T cell suppression. In addition, PI3K/AKT/mTOR and Wnt/β-catenin pathways promote chemoresistance through metabolic adaptation. By elucidating the link between metabolic reprogramming and immune evasion, this study identifies key metabolic vulnerabilities and potential drug targets in ovarian cancer. Our findings support the development of metabolically targeted therapies and increase the utility of organoid-based precision medicine models.
Collapse
Affiliation(s)
- Lanyue Zhang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jiangnan Zhao
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Chunyu Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jianxi Wu
- Department of Preventive Medicine, Southwest Medical University, Luzhou, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Qin Wang
- Sichuan Provincial Center for Gynecology and Breast Diseases (Gynecology), Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Shah DD, Chorawala MR, Raghani NR, Patel R, Fareed M, Kashid VA, Prajapati BG. Tumor microenvironment: recent advances in understanding and its role in modulating cancer therapies. Med Oncol 2025; 42:117. [PMID: 40102282 DOI: 10.1007/s12032-025-02641-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Tumor microenvironment (TME) denotes the non-cancerous cells and components presented in the tumor, including molecules produced and released by them. Interactions between cancer cells, immune cells, stromal cells, and the extracellular matrix within the TME create a dynamic ecosystem that can either promote or hinder tumor growth and spread. The TME plays a pivotal role in either promoting or inhibiting tumor growth and dissemination, making it a critical factor to consider in the development of effective cancer therapies. Understanding the intricate interplay within the TME is crucial for devising effective cancer therapies. Combination therapies involving inhibitors of immune checkpoint blockade (ICB), and/or chemotherapy now offer new approaches for cancer therapy. However, it remains uncertain how to best utilize these strategies in the context of the complex tumor microenvironment. Oncogene-driven changes in tumor cell metabolism can impact the TME to limit immune responses and present barriers to cancer therapy. Cellular and acellular components in tumor microenvironment can reprogram tumor initiation, growth, invasion, metastasis, and response to therapies. Components in the TME can reprogram tumor behavior and influence responses to treatments, facilitating immune evasion, nutrient deprivation, and therapeutic resistance. Moreover, the TME can influence angiogenesis, promoting the formation of blood vessels that sustain tumor growth. Notably, the TME facilitates immune evasion, establishes a nutrient-deprived milieu, and induces therapeutic resistance, hindering treatment efficacy. A paradigm shift from a cancer-centric model to a TME-centric one has revolutionized cancer research and treatment. However, effectively targeting specific cells or pathways within the TME remains a challenge, as the complexity of the TME poses hurdles in designing precise and effective therapies. This review highlights challenges in targeting the tumor microenvironment to achieve therapeutic efficacy; explore new approaches and technologies to better decipher the tumor microenvironment; and discuss strategies to intervene in the tumor microenvironment and maximize therapeutic benefits.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India.
| | - Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar, Gujarat, 382355, India
| | - Rajanikant Patel
- Department of Product Development, Granules Pharmaceuticals Inc., 3701 Concorde Parkway, Chantilly, VA, 20151, USA
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, 13713, Riyadh, Saudi Arabia
| | - Vivekanand A Kashid
- MABD Institute of Pharmaceutical Education and Research, Babhulgaon, Yeola, Nashik, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Kherva, Mehsana, Gujarat, 384012, India.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
13
|
Xiao Z, Puré E. The fibroinflammatory response in cancer. Nat Rev Cancer 2025:10.1038/s41568-025-00798-8. [PMID: 40097577 DOI: 10.1038/s41568-025-00798-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 03/19/2025]
Abstract
Fibroinflammation refers to the highly integrated fibrogenic and inflammatory responses mediated by the concerted function of fibroblasts and innate immune cells in response to tissue perturbation. This process underlies the desmoplastic remodelling of the tumour microenvironment and thus plays an important role in tumour initiation, growth and metastasis. More specifically, fibroinflammation alters the biochemical and biomechanical signalling in malignant cells to promote their proliferation and survival and further supports an immunosuppressive microenvironment by polarizing the immune status of tumours. Additionally, the presence of fibroinflammation is often associated with therapeutic resistance. As such, there is increasing interest in targeting this process to normalize the tumour microenvironment and thus enhance the treatment of solid tumours. Herein, we review advances made in unravelling the complexity of cancer-associated fibroinflammation that can inform the rational design of therapies targeting this.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Zhang Y, Chen H, Mo H, Zhao N, Sun X, Liu B, Gao R, Xu B, Zhang Z, Liu Z, Ma F. Distinct cellular mechanisms underlie chemotherapies and PD-L1 blockade combinations in triple-negative breast cancer. Cancer Cell 2025; 43:446-463.e7. [PMID: 39919737 DOI: 10.1016/j.ccell.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 11/05/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025]
Abstract
Combining immune checkpoint blockade (ICB) with chemotherapy shows promise for treating triple-negative breast cancer (TNBC), though the mechanisms remain incompletely understood. Here, we integrate published and new single-cell RNA sequencing (scRNA-seq) data to investigate the tumor immune microenvironment (TIME) in TNBC patients treated with paclitaxel (PTX), nab-paclitaxel (Nab-PTX), and their combinations with the anti-PD-L1 antibody atezolizumab (ATZ). Compared to ATZ plus PTX, ATZ plus Nab-PTX rewires TCF7+ stem-like effector memory CD8+ T cells (Tsem) and CD4+ T follicular helper (Tfh) cells. Nab-paclitaxel, unlike PTX, also reshapes the myeloid compartment, expanding mast cells and pro-inflammatory macrophages. Our analyses in human TNBC and murine models underscore the crucial role of mast cells in orchestrating anti-tumor immune responses, likely by promoting the recruitment and activation of T and B cells. In vivo experiments demonstrate that activating mast cells alongside PD-L1 blockade attenuates TNBC progression, suggesting mast cells as a promising adjunct for enhancing ICB therapy efficacy.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China.
| | - Hongyan Chen
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hongnan Mo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ning Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaoying Sun
- Department of Medical Oncology, Cancer Hospital of HuanXing, ChaoYang District, Beijing 100005, China
| | - Baolin Liu
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ranran Gao
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China
| | - Binghe Xu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Zemin Zhang
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China.
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Fei Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
15
|
Wei X, He Y, Yu Y, Tang S, Liu R, Guo J, Jiang Q, Zhi X, Wang X, Meng D. The Multifaceted Roles of BACH1 in Disease: Implications for Biological Functions and Therapeutic Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412850. [PMID: 39887888 PMCID: PMC11905017 DOI: 10.1002/advs.202412850] [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: 10/13/2024] [Revised: 12/22/2024] [Indexed: 02/01/2025]
Abstract
BTB domain and CNC homolog 1 (BACH1) belongs to the family of basic leucine zipper proteins and is expressed in most mammalian tissues. It can regulate its own expression and play a role in transcriptionally activating or inhibiting downstream target genes. It has a crucial role in various biological processes, such as oxidative stress, cell cycle, heme homeostasis, and immune regulation. Recent research highlights BACH1's significant regulatory roles in a series of conditions, including stem cell pluripotency maintenance and differentiation, growth, senescence, and apoptosis. BACH1 is closely associated with cardiovascular diseases and contributes to angiogenesis, atherosclerosis, restenosis, pathological cardiac hypertrophy, myocardial infarction, and ischemia/reperfusion (I/R) injury. BACH1 promotes tumor cell proliferation and metastasis by altering tumor metabolism and the epithelial-mesenchymal transition phenotype. Moreover, BACH1 appears to show an adverse role in diseases such as neurodegenerative diseases, gastrointestinal disorders, leukemia, pulmonary fibrosis, and skin diseases. Inhibiting BACH1 may be beneficial for treating these diseases. This review summarizes the role of BACH1 and its regulatory mechanism in different cell types and diseases, proposing that precise targeted intervention of BACH1 may provide new strategies for human disease prevention and treatment.
Collapse
Affiliation(s)
- Xiangxiang Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Yunquan He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Yueyang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Sichong Tang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Ruiwen Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Jieyu Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Qingjun Jiang
- Department of Vascular & Endovascular Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Xiuling Zhi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Xinhong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Dan Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| |
Collapse
|
16
|
Kertmen N, Kavgaci G, Koc I, Sagol SP, Isikay AI, Yazici G. Sequential immunotherapy and bevacizumab treatments in glioblastoma multiforme: A case series and review of the literature. Oncol Lett 2025; 29:146. [PMID: 39877061 PMCID: PMC11773301 DOI: 10.3892/ol.2025.14892] [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: 06/18/2024] [Accepted: 12/31/2024] [Indexed: 01/31/2025] Open
Abstract
Glioblastoma multiforme (GBM) is a tumor with a high refractory rate to immunotherapy and a low tumor mutational burden phenotype, leading to limited immunogenic neoantigens. The present study aimed to investigate the sequential use of immunotherapy and bevacizumab in patients with GBM, exploring the clinical outcomes and potential complications. Patients received various combinations of immunotherapy and bevacizumab after standard treatment, including surgery, radiotherapy and temozolomide. Clinical courses, radiological findings and treatment outcomes were monitored and documented during each clinical visit through routine physical examinations, imaging studies and review of medical records. The efficacy and side effects of this sequential drug approach remained unclear. The common features of these patients were a marked decline in cognitive function and clinical deterioration, assessed clinically in the absence of obvious tumor progression. Radiological evaluation was also performed, particularly for possible cerebrovascular events. In these cases, the potential for sequential treatment to suppress tumors while inducing cerebrovascular events was also investigated, and patients were not lost to overt tumor progression. Notably, further research is required to clarify the mechanisms of action and complications associated with the sequential use of immunotherapy and bevacizumab in the treatment of GBM.
Collapse
Affiliation(s)
- Neyran Kertmen
- Department of Medical Oncology, Hacettepe University Faculty of Medicine, Ankara, Ankara 06230, Turkey
| | - Gozde Kavgaci
- Department of Medical Oncology, Hacettepe University Faculty of Medicine, Ankara, Ankara 06230, Turkey
| | - Ilgin Koc
- Department of Medical Oncology, Hacettepe University Faculty of Medicine, Ankara, Ankara 06230, Turkey
| | - Safak Parlak Sagol
- Department of Radiology, Hacettepe University Faculty of Medicine, Ankara, Ankara 06230, Turkey
| | - Ahmet Ilkay Isikay
- Department of Neurosurgery, Hacettepe University Faculty of Medicine, Ankara, Ankara 06230, Turkey
| | - Gozde Yazici
- Department of Radiation Oncology, Hacettepe University Faculty of Medicine, Ankara, Ankara 06230, Turkey
| |
Collapse
|
17
|
Wang X, Song J, Qiu Q, Su Y, Wang L, Cao X. A Stacked Multimodality Model Based on Functional MRI Features and Deep Learning Radiomics for Predicting the Early Response to Radiotherapy in Nasopharyngeal Carcinoma. Acad Radiol 2025; 32:1631-1644. [PMID: 39496536 DOI: 10.1016/j.acra.2024.10.011] [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: 09/03/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND This study aimed to construct and assess a comprehensive model that integrates MRI-derived deep learning radiomics, functional imaging (fMRI), and clinical indicators to predict early efficacy of radiotherapy in nasopharyngeal carcinoma (NPC). METHODS This retrospective study recruited NPC patients with radiotherapy from two Chinese hospitals between October 2018 and July 2022, divided into a training set (hospital I, 194 cases), an internal validation set (hospital I, 82 cases), and an external validation set (hospital II, 40 cases). We extracted 3404 radiomic features and 2048 deep learning features from multi-sequence MRI includes T1WI, CE-T1WI, T2WI and T2WI/FS. Additionally, both the Apparent diffusion coefficient (ADC), its maximum (ADCmax) and Tumor blood flow (TBF), its maximum (TBFmax) were obtained by Diffusion-weighted imaging (DWI) and Arterial spin labeling (ASL) respectively. We used four classifiers (LR, XGBoost, SVM and KNN) and stacked algorithm as model construction methods. The area under the receiver operating characteristic curve (AUC) and decision curve analysis was used to assess models. RESULTS The manual radiomics model based on XGBoost and the deep learning model based on KNN (the AUCs in the training set: 0.909, 0.823, respectively) showed better predictive efficacy than other machine learning algorithms. The stacked model that integrated MRI-based deep learning radiomics, fMRI, and hematological indicators, has the strongest efficacy prediction ability of AUC in the training set [0.984 (95%CI: 0.972-0.996)], the internal validation set [0.936 (95%CI: 0.885-0.987)], and the external validation set [0.959 (95%CI: 0.901-1.000)]. CONCLUSION Our research has developed a clinical-radiomics integrated model based on MRI which can predict early radiotherapy response in NPC and provide guidance for personalized treatment.
Collapse
Affiliation(s)
- Xiaowen Wang
- Shandong University Cancer Center, Jinan, Shandong, China (X.W.); Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (X.W., X.C.)
| | - Jian Song
- Medical Imageology, Shandong Medical College, Jinan, China (J.S.)
| | - Qingtao Qiu
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Q.Q., Y.S., L.W.)
| | - Ya Su
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Q.Q., Y.S., L.W.)
| | - Lizhen Wang
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Q.Q., Y.S., L.W.)
| | - Xiujuan Cao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (X.W., X.C.).
| |
Collapse
|
18
|
Chu B, Chen Y, Pan J. Prognostic significance of systemic immune inflammation index for ovarian cancer: An updated systematic review and meta-analysis. J Ovarian Res 2025; 18:41. [PMID: 40016853 PMCID: PMC11869409 DOI: 10.1186/s13048-025-01626-1] [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: 02/14/2025] [Indexed: 03/01/2025] Open
Abstract
OBJECTIVE Several inflammatory indices have been used to assess the prognosis of ovarian cancer, with variable results. This review assessed whether the systemic immune inflammation index (SII) can predict outcomes in patients with ovarian cancer. METHODS Embase, PubMed, CENTRAL, Web of Science, and Scopus databases were searched by the two reviewers from inception to 15th October 2024 for studies assessing the relationship between SII and overall survival (OS) or disease-free survival (DFS). RESULTS Ten studies with eleven cohorts were included. Pooled analysis showed that higher SII was a significant predictor of poor OS (HR: 2.35 95% CI: 1.56, 3.55 I2 = 88%) and worse DFS (HR: 2.51 95% CI: 1.71, 3.67 I2 = 80%) after ovarian cancer. Sensitivity analysis failed to change the significance of the results. No publication bias was noted. Most results remained significant on subgroup analyses based on location, sample size, FIGO stage, treatment, adjusted outcomes, cut-off of SII, method of determining cut-off, and quality score. CONCLUSIONS SII can be a potential predictor of OS and DFS after ovarian cancer. Further studies are required to improve the evidence.
Collapse
Affiliation(s)
- Boliang Chu
- Department of Gynecology, Huzhou Maternity & Child Health Care Hospital, No. 2, East Street, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Yingying Chen
- Department of Gynecology, Huzhou Maternity & Child Health Care Hospital, No. 2, East Street, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Jiewei Pan
- Department of Gynecology, Huzhou Maternity & Child Health Care Hospital, No. 2, East Street, Wuxing District, Huzhou, Zhejiang Province, 313000, China.
| |
Collapse
|
19
|
Misawa K, Bhat H, Adusumilli PS, Hou Z. Combinational CAR T-cell therapy for solid tumors: Requisites, rationales, and trials. Pharmacol Ther 2025; 266:108763. [PMID: 39617146 PMCID: PMC11848936 DOI: 10.1016/j.pharmthera.2024.108763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved potent antitumor efficacy in hematological malignancies; however, because of limitations in CAR T-cell recruitment, infiltration, activation, and functional persistence in the tumor, its efficacy in solid tumors has been suboptimal. To overcome these challenges, combinational strategies that include chemotherapy, radiation therapy, or immune checkpoint inhibitor agent therapy with CAR T-cell therapy are being investigated. The established functional characteristics of the abovementioned therapies provide a rationale for the use of a combinational approach with CAR T cells. Chemotherapy reshapes the peritumoral stroma, decreases the immunosuppressive cell population, and promotes a proinflammatory milieu, all of which allow for increased recruitment, infiltration, and accumulation of CAR T cells. Radiation therapy promotes a chemokine gradient, which augments tumor infiltration by CAR T cells and further increases expression of tumor-associated antigens, allowing for increased activation of CAR T cells. Immune checkpoint inhibitor agent therapy inactivates T-cell exhaustion pathways-most notably, the PD1/PDL1 pathway-thereby improving the functional persistence of CAR T cells and promoting endogenous immunity. In this review, we discuss the requisites and rationales for combinational therapy, and we review 25 ongoing phase I and II clinical trials, of which 4 use chemotherapy, 3 use radiation therapy, 11 use immunotherapy, and 7 use another agent. While safety, efficacy, and improved outcomes are the primary goals of these ongoing studies, the knowledge gained from them will help pave the way for subsequent studies focused on optimizing combinational regimens and identifying predictive biomarkers.
Collapse
Affiliation(s)
- Kyohei Misawa
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Hina Bhat
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Zhaohua Hou
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
20
|
Sakai SA, Nomura R, Nagasawa S, Chi S, Suzuki A, Suzuki Y, Imai M, Nakamura Y, Yoshino T, Ishikawa S, Tsuchihara K, Kageyama SI, Yamashita R. SpatialKNifeY (SKNY): Extending from spatial domain to surrounding area to identify microenvironment features with single-cell spatial omics data. PLoS Comput Biol 2025; 21:e1012854. [PMID: 39965034 PMCID: PMC11849985 DOI: 10.1371/journal.pcbi.1012854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/24/2025] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
Single-cell spatial omics analysis requires consideration of biological functions and mechanisms in a microenvironment. However, microenvironment analysis using bioinformatic methods is limited by the need to detect histological morphology and extend it to the surrounding area. In this study, we developed SpatialKNifeY (SKNY), an image-processing-based toolkit that detects spatial domains that potentially reflect histology and extends these domains to the microenvironment. Using spatial transcriptomic data from breast cancer, we applied the SKNY algorithm to identify tumor spatial domains, followed by clustering of the domains, trajectory estimation, and spatial extension to the tumor microenvironment (TME). The results of the trajectory estimation were consistent with the known mechanisms of cancer progression. We observed tumor vascularization and immunodeficiency at mid- and late-stage progression in TME. Furthermore, we applied the SKNY to integrate and cluster the spatial domains of 14 patients with metastatic colorectal cancer, and the clusters were divided based on the TME characteristics. In conclusion, the SKNY facilitates the determination of the functions and mechanisms in the microenvironment and cataloguing of the features.
Collapse
Affiliation(s)
- Shunsuke A. Sakai
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Ryosuke Nomura
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Satoi Nagasawa
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
- Department of Breast Surgery, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - SungGi Chi
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Ayako Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Mitsuho Imai
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Chiba, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Takayuki Yoshino
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Division of Pathology, National Cancer Center Exploratory Oncology Research & Clinical Trial Center, Kashiwa, Chiba, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Shun-Ichiro Kageyama
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| |
Collapse
|
21
|
To A, Yu Z, Sugimura R. Recent advancement in the spatial immuno-oncology. Semin Cell Dev Biol 2025; 166:22-28. [PMID: 39705969 DOI: 10.1016/j.semcdb.2024.12.003] [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: 06/21/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
Recent advancements in spatial transcriptomics and spatial proteomics enabled the high-throughput profiling of single or multi-cell types and cell states with spatial information. They transformed our understanding of the higher-order architectures and paired cell-cell interactions within a tumor microenvironment (TME). Within less than a decade, this rapidly emerging field has discovered much crucial fundamental knowledge and significantly improved clinical diagnosis in the field of immuno-oncology. This review summarizes the conceptual frameworks to understand spatial omics data and highlights the updated knowledge of spatial immuno-oncology.
Collapse
Affiliation(s)
- Alex To
- School of Biomedical Sciences, University of Hong Kong, Hong Kong
| | - Zou Yu
- School of Biomedical Sciences, University of Hong Kong, Hong Kong
| | - Ryohichi Sugimura
- School of Biomedical Sciences, University of Hong Kong, Hong Kong; Centre for Translational Stem Cell Biology, Hong Kong.
| |
Collapse
|
22
|
Zhu Y, Chen J, Chen C, Tang R, Xu J, Shi S, Yu X. Deciphering mechanical cues in the microenvironment: from non-malignant settings to tumor progression. Biomark Res 2025; 13:11. [PMID: 39849659 PMCID: PMC11755887 DOI: 10.1186/s40364-025-00727-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/05/2025] [Indexed: 01/25/2025] Open
Abstract
The tumor microenvironment functions as a dynamic and intricate ecosystem, comprising a diverse array of cellular and non-cellular components that precisely orchestrate pivotal tumor behaviors, including invasion, metastasis, and drug resistance. While unraveling the intricate interplay between the tumor microenvironment and tumor behaviors represents a tremendous challenge, recent research illuminates a crucial biological phenomenon known as cellular mechanotransduction. Within the microenvironment, mechanical cues like tensile stress, shear stress, and stiffness play a pivotal role by activating mechanosensitive effectors such as PIEZO proteins, integrins, and Yes-associated protein. This activation initiates cascades of intrinsic signaling pathways, effectively linking the physical properties of tissues to their physiological and pathophysiological processes like morphogenesis, regeneration, and immunity. This mechanistic insight offers a novel perspective on how the mechanical cues within the tumor microenvironment impact tumor behaviors. While the intricacies of the mechanical tumor microenvironment are yet to be fully elucidated, it exhibits distinct physical attributes from non-malignant tissues, including elevated solid stresses, interstitial hypertension, augmented matrix stiffness, and enhanced viscoelasticity. These traits exert notable influences on tumor progression and treatment responses, enriching our comprehension of the multifaceted nature of the microenvironment. Through this innovative review, we aim to provide a new lens to decipher the mechanical attributes within the tumor microenvironment from non-malignant contexts, broadening our knowledge on how these factors promote or inhibit tumor behaviors, and thus offering valuable insights to identify potential targets for anti-tumor strategies.
Collapse
Affiliation(s)
- Yicheng Zhu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jiaoshun Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chen Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
23
|
Geisler L, Detjen K, Hellberg T, Kohlhepp M, Grötzinger C, Knorr J, Eichhorn I, Mohr R, Holtmann T, Wiedenmann B, Tacke F, Roderburg C, Wree A. miR-223 and Chromogranin A Affect Inflammatory Immune Cell Activation in Liver Metastasis of Neuroendocrine Neoplasms. Cells 2025; 14:111. [PMID: 39851539 PMCID: PMC11763622 DOI: 10.3390/cells14020111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Neuroendocrine neoplasms (NENs) are a diverse group originating from endocrine cells/their precursors in pancreas, small intestine, or lung. The key serum marker is chromogranin A (CgA). While commonly elevated in patients with NEN, its prognostic value is still under discussion. Secretion/posttranslational proteolytic cleavage of CgA results in multiple bioactive fragments, which are essential regulators of the cardiovascular and immune system. miR-223, regulator of Nrlp3 inflammasome and neutrophil activation, was recently found to have decreased in patients with NEN. We performed flow cytometry of circulating neutrophils in a patient cohort (n = 10) with NEN, microdissection and histology of tumor tissue. Subsequently, in vitro transfections using the well-established human pancreatic NEN cell line (BON), and co-culture experiments with primary macrophages and neutrophils were performed. Serum miR-223 in patients correlated with the expression of the neutrophil activation marker CD15 in circulating cells. Neutrophilic CD62L/CD63 showed good discrimination compared to healthy controls. Immune cell-derived miR-155, miR-193 and miR-223 colocalize with neutrophil in the extra-tumoral tissue alongside Nlrp3-associated caspase-1 activation. miR-223 knockdown in BON decreased the CgA intracellularly, increased in cellular granularity and caspase-1 activation. Plasmin inhibitor a2-aP reverted those effects. Western Blot showed fragmented CgA following miR-223 knockdown, which altered the inflammatory potential of neutrophils. Our data hence provide initial insights into an immunoregulatory mechanism via miR-223 and CgA in NEN cells, as regulation of miR-223 in NEN may affect tumor-associated inflammation.
Collapse
Affiliation(s)
- Lukas Geisler
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
- Department of Biology, Humboldt University of Berlin, 10099 Berlin, Germany
| | - Katharina Detjen
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Teresa Hellberg
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Marlene Kohlhepp
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Carsten Grötzinger
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Jana Knorr
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Ines Eichhorn
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Raphael Mohr
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Theresa Holtmann
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Bertram Wiedenmann
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
| | - Alexander Wree
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, 13353 Berlin, Germany
| |
Collapse
|
24
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [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: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
25
|
Liang C, Wang Y, Zhao R, Du J, Yao J, Khan AUR, Zhu Y, Xia H, Zhu T. Multifunctional hybrid poly(ester-urethane)urea/resveratrol electrospun nanofibers for a potential vascularizing matrix. SOFT MATTER 2025; 21:55-67. [DOI: doi:10.1039/d4sm00937a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
P/R-1.0 nanofiber with excellent antioxidant, blood and cell compatibility fibricated via electrospinning for a potential vascularizing matrix.
Collapse
Affiliation(s)
- Chen Liang
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, P. R. China
| | - Yanan Wang
- Department of Minimally Invasive Spine Surgery, Shandong Wendeng Orthopedic Hospital, 1 Fengshan Rd., Weihai 264400, Shandong, P. R. China
| | - Renliang Zhao
- Orthopedics Research Institute, Trauma Medical Center, Department of Orthopedics, West China Hospital, Sichuan University, 37 Guoxue Ln., Chengdu 610041, Sichuan, P. R. China
| | - Juan Du
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, P. R. China
| | - Jin Yao
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, P. R. China
| | - Atta ur Rehman Khan
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, P. R. China
| | - Youwei Zhu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd., Shanghai 200025, P. R. China
- Shanghai Key Laboratory of Pancreatic Neoplasms Translational Medicine, 197 Ruijin 2nd Rd., Shanghai 200025, P. R. China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd., Shanghai 200025, P. R. China
| | - Huitang Xia
- Department of Plastic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, 16766 Jingshi Rd., Jinan 250014, Shandong, P. R. China
- Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, 16766 Jingshi Rd., Jinan 250014, Shangdong, P. R. China
| | - Tonghe Zhu
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, P. R. China
| |
Collapse
|
26
|
Furube T, Nakashima D, Matsuda S, Mikami K, Hatakeyama T, Takeuchi M, Fukuda K, Ueno A, Okita H, Kawakubo H, Nakamura M, Nagura T, Kitagawa Y. Evaluating stiffness of gastric wall using laser resonance frequency analysis for gastric cancer. Cancer Sci 2025; 116:143-151. [PMID: 39468628 PMCID: PMC11711037 DOI: 10.1111/cas.16383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
Tumor stiffness is drawing attention as a novel axis that is orthogonal to existing parameters such as pathological examination. We developed a new diagnostic method that focuses on differences in stiffness between tumor and normal tissue. This study comprised a clinical application of laser resonance frequency analysis (L-RFA) for diagnosing gastric cancer. L-RFA allows for precise and contactless evaluation of tissue stiffness. We used a laser to create vibrations on a sample's surface that were then measured using a vibrometer. The data were averaged and analyzed to enhance accuracy. In the agarose phantom experiments, a clear linear correlation was observed between the Young's modulus of the phantoms (0.34-0.71 MPa) and the summation of normalized vibration peaks (Score) in the 1950-4050 Hz range (R2 = 0.93145). Higher Young's moduli also resulted in lower vibration peaks at the excitation frequency, signal-to-noise (S/N) ratios, and harmonic peaks. We also conducted L-RFA measurements on gastric cancer specimens from two patients who underwent robot-assisted distal gastrectomy. Our results revealed significant waveform differences between tumor and normal regions, similar to the findings in agarose phantoms, with tumor regions exhibiting lower vibration peaks at the excitation frequency, S/N ratios, and harmonic peaks. Statistical analysis confirmed significant differences in the score between normal and tumor regions (p = 0.00354). L-RFA was able to assess tumor stiffness and holds great promise as a noninvasive diagnostic tool for gastric cancer.
Collapse
Affiliation(s)
- Tasuku Furube
- Department of SurgeryKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Daisuke Nakashima
- Department of Orthopaedic SurgeryKeio University School of MedicineShinjuku‐kuTokyoJapan
- Department of Clinical BiomechanicsKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Satoru Matsuda
- Department of SurgeryKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Katsuhiro Mikami
- Faculty of Biology‐Oriented Science and TechnologyKindai UniversityKinokawaWakayamaJapan
| | - Takuto Hatakeyama
- Department of Orthopaedic SurgeryKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Masashi Takeuchi
- Department of SurgeryKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Kazumasa Fukuda
- Department of SurgeryKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Akihisa Ueno
- Division of Diagnostic PathologyKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Hajime Okita
- Division of Diagnostic PathologyKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Hirofumi Kawakubo
- Department of SurgeryKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Masaya Nakamura
- Department of Orthopaedic SurgeryKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Takeo Nagura
- Department of Orthopaedic SurgeryKeio University School of MedicineShinjuku‐kuTokyoJapan
- Department of Clinical BiomechanicsKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Yuko Kitagawa
- Department of SurgeryKeio University School of MedicineShinjuku‐kuTokyoJapan
| |
Collapse
|
27
|
Yang J, Wang H, Li B, Liu J, Zhang X, Wang Y, Peng J, Gao L, Wang X, Hu S, Zhang W, Hong L. Inhibition of ACSS2 triggers glycolysis inhibition and nuclear translocation to activate SIRT1/ATG5/ATG2B deacetylation axis, promoting autophagy and reducing malignancy and chemoresistance in ovarian cancer. Metabolism 2025; 162:156041. [PMID: 39362518 DOI: 10.1016/j.metabol.2024.156041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Metabolic reprogramming is a hallmark of cancer, characterized by a high dependence on glycolysis and an enhanced utilization of acetate as an alternative carbon source. ACSS2 is a critical regulator of acetate metabolism, playing a significant role in the development and progression of various malignancies. ACSS2 facilitates the conversion of acetate to acetyl-CoA, which participates in multiple metabolic pathways and functions as an epigenetic regulator of protein acetylation, thereby modulating key cellular processes such as autophagy. However, the roles and intrinsic connections of ACSS2, glycolysis, protein acetylation, and autophagy in ovarian cancer (OC) remain to be elucidated. BASIC PROCEDURES Utilizing clinical specimens and online databases, we analysed the expression of ACSS2 in OC and its relationship with clinical prognosis. By knocking down ACSS2, we evaluated its effects on the malignant phenotype, acetate metabolism, glycolysis, and autophagy. The metabolic alterations in OC cells were comprehensively analysed using Seahorse assays, transmission electron microscopy, membrane potential measurements, and stable-isotope labeling techniques. CUT&TAG and co-immunoprecipitation techniques were employed to explore the deacetylation of autophagy-related proteins mediated by ACSS2 via SIRT1. Additionally, through molecular docking, transcriptome sequencing, and metabolomics analyses, we validated the pharmacological effects of paeonol on ACSS2 and the glycolytic process in OC cells. Finally, both in vitro and in vivo experiments were performed to investigate the impact of paeonol on autophagy and its anti-OC effects mediated through the ACSS2/SIRT1 deacetylation axis. MAIN FINDINGS ACSS2 is significantly upregulated in OC and is associated with poor prognosis. Knockdown of ACSS2 inhibits OC cells proliferation, migration, invasion, angiogenesis, and platinum resistance, while reducing tumour burden in vivo. Mechanistically, inhibiting ACSS2 reduces acetate metabolism and suppresses glycolysis by targeting HXK2. This glycolytic reduction promotes the translocation of ACSS2 from the cytoplasm to the nucleus, leading to increased expression of the deacetylase SIRT1. SIRT1 mediates the deacetylation of autophagy-related proteins, such as ATG5 and ATG2B, thereby significantly activating autophagy in OC cells and exerting antitumor effects. Paeonol inhibits acetate metabolism and glycolysis in OC cells by targeting ACSS2. Paeonol activates autophagy through the ACSS2/SIRT1/ATG5/ATG2B deacetylation axis, demonstrating inhibition of OC in vitro and in vivo. PRINCIPAL CONCLUSIONS Pae can serve as an effective, low-toxicity, multi-targeted drug targeting ACSS2 and glycolysis. It activates autophagy through the ACSS2/SIRT1/ATG5/ATG2B deacetylation signalling cascade, thereby exerting anti-OC effects. Our study provides new insights into the malignant mechanisms of OC and offers a novel strategy for its treatment.
Collapse
Affiliation(s)
- Jiang Yang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China; Department of Obstetrics and Gynaecology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, PR China
| | - Haoyu Wang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Bingshu Li
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Jingchun Liu
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Xiaoyi Zhang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Ying Wang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Jiaxin Peng
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Likun Gao
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Xinqi Wang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Siyuan Hu
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Wenyi Zhang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Li Hong
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China.
| |
Collapse
|
28
|
Lin QX, Song WW, Xie WX, Deng YT, Gong YN, Liu YR, Tian Y, Zhao WY, Tian L, Gu DN. Sequential treatment of anti-PD-L1 therapy prior to anti-VEGFR2 therapy contributes to more significant clinical benefits in non-small cell lung cancer. Neoplasia 2025; 59:101077. [PMID: 39561585 PMCID: PMC11617296 DOI: 10.1016/j.neo.2024.101077] [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: 07/18/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024]
Abstract
OBJECTIVE Anti-angiogenic therapy and immune checkpoint blockade therapy are currently important treatments for non-small cell lung cancer. However, the combined use of the two therapies is controversial, and few studies have investigated the effects of different time sequences of the two therapies on treatment outcomes. METHODS The tumor-bearing mouse model was established and the mice were divided into four groups, including AA-ICB sequence group, ICB-AA sequence group, synchronization group and the control group. Immunohistochemistry was used to assess tumor microvessels and PD-L1 expression. Selected immune cell populations were evaluated using flow cytometry. Meta-analysis and clinical information were used to elucidate the clinical effects of administration sequence. RESULTS We found that anti-PD-L1 treatment followed by anti-VEGFR2 therapy exerts the best inhibitory effect on tumor growth. Different sequences of anti-angiogenic therapy and immune checkpoint blockade therapy resulted in different proportions of tumor microvessels and immune cell populations in the tumor microenvironment. We further revealed that the administration of anti-PD-L1 before anti-VEGFR brought more normalized tumor blood vessels and CD8+T cell infiltration and reduced immunosuppressive cells in the tumor microenvironment. Subsequent re-transplantation experiments confirmed the long-term benefits of this treatment strategy. The meta-analysis reinforced that immunotherapy prior to anti-angiogenic therapy or combination therapy have better therapeutic effects in advanced non-small cell lung cancer. CONCLUSION Our study demonstrated that the therapeutic effect of anti-angiogenic treatment after immune checkpoint therapy was superior to that of concurrent therapy, whereas anti-angiogenic therapy followed by immunotherapy did not bring more significant clinical benefits than independent monotherapy.
Collapse
Affiliation(s)
- Qiao-Xin Lin
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wen-Wen Song
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wen-Xia Xie
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi-Ting Deng
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan-Na Gong
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi-Ru Liu
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Ya Zhao
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dian-Na Gu
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
29
|
Guo Z, Zhu Z, Lin X, Wang S, Wen Y, Wang L, Zhi L, Zhou J. Tumor microenvironment and immunotherapy for triple-negative breast cancer. Biomark Res 2024; 12:166. [PMID: 39741315 DOI: 10.1186/s40364-024-00714-6] [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: 10/16/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer known for its high aggressiveness and poor prognosis. Conventional treatment of TNBC is challenging due to its heterogeneity and lack of clear targets. Recent advancements in immunotherapy have shown promise in treating TNBC, with immune checkpoint therapy playing a significant role in comprehensive treatment plans. The tumor microenvironment (TME), comprising immune cells, stromal cells, and various cytokines, plays a crucial role in TNBC progression and response to immunotherapy. The high presence of tumor-infiltrating lymphocytes and immune checkpoint proteins in TNBC indicates the potential of immunotherapeutic strategies. However, the complexity of the TME, while offering therapeutic targets, requires further exploration of its multiple roles in immunotherapy. In this review, we discuss the interaction mechanism between TME and TNBC immunotherapy based on the characteristics and composition of TME, and elaborate on and analyze the effect of TME on immunotherapy, the potential of TME as an immune target, and the ability of TME as a biomarker. Understanding these dynamics will offer new insights for enhancing therapeutic approaches and investigating stratification and prognostic markers for TNBC patients.
Collapse
Affiliation(s)
- Zijie Guo
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Ziyu Zhu
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Xixi Lin
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Shenkangle Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Yihong Wen
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Linbo Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Lili Zhi
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Jichun Zhou
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
30
|
Feng C, Wang Y, Song W, Liu T, Mo H, Liu H, Wu S, Qin Z, Wang Z, Tao Y, He L, Tang S, Xie Y, Wang Q, Li T. Spatially-resolved analyses of muscle invasive bladder cancer microenvironment unveil a distinct fibroblast cluster associated with prognosis. Front Immunol 2024; 15:1522582. [PMID: 39759522 PMCID: PMC11695344 DOI: 10.3389/fimmu.2024.1522582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/05/2024] [Indexed: 01/07/2025] Open
Abstract
Background Muscle-invasive bladder cancer (MIBC) is a prevalent cancer characterized by molecular and clinical heterogeneity. Assessing the spatial heterogeneity of the MIBC microenvironment is crucial to understand its clinical significance. Methods In this study, we used imaging mass cytometry (IMC) to assess the spatial heterogeneity of MIBC microenvironment across 185 regions of interest in 40 tissue samples. We focused on three primary parameters: tumor (T), leading-edge (L), and nontumor (N). Cell gating was performed using the Cytobank platform. We calculated the Euclidean distances between cells to determine cellular interactions and performed single-cell RNA sequencing (scRNA-seq) to explore the molecular characteristics and mechanisms underlying specific fibroblast (FB) clusters. scRNA-seq combined with spatial transcriptomics (ST) facilitated the identification of ligand-receptor (L-R) pairs that mediate interactions between specific FB clusters and endothelial cells. Machine learning algorithms were used to construct a prognostic gene signature. Results The microenvironments in the N, L, and T regions of MIBC exhibited spatial heterogeneity and regional diversity in their components. A distinct FB cluster located in the L region-identified as S3-is strongly associated with poor prognosis. IMC analyses demonstrated a close spatial association between S3 and endothelial cells, with S3-positive tumors exhibiting increased blood vessel density and altered vascular morphology. The expression of vascular endothelial growth factor receptor and active vascular sprouting were significant in S3-positive tumors. scRNA-seq and ST analyses indicated that the genes upregulated in S3 were associated with angiogenesis. NOTCH1-JAG2 signaling pathway was identified as a significant L-R pair specific to S3 and endothelial cell interactions. Further analysis indicated that YAP1 was a potential regulator of S3. Machine learning algorithms and Gene Set Variation Analysis were used to establish an S3-related gene signature that was associated with the poor prognosis of tumors including MIBC, mesothelioma, glioblastoma multiforme, lower-grade glioma, stomach adenocarcinoma, uveal melanoma, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, and lung squamous cell carcinoma. Conclusions We assessed the spatial landscape of the MIBC microenvironment and revealed a specific FB cluster with prognostic potential. These findings offer novel insights into the spatial heterogeneity of the MIBC microenvironment and highlight its clinical significance.
Collapse
Affiliation(s)
- Chao Feng
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yaobang Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wuyue Song
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tao Liu
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Han Mo
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hui Liu
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shulin Wu
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zezu Qin
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhenxing Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yuting Tao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Liangyu He
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shaomei Tang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Yuanliang Xie
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Qiuyan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Tianyu Li
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
31
|
Hanada K, Saito Y, Takagi T, Go M, Nakano Y, Inagawa T, Hirai H, Fruttiger M, Itoh S, Itoh F. Reduced lung metastasis in endothelial cell-specific transforming growth factor β type II receptor-deficient mice with decreased CD44 expression. iScience 2024; 27:111502. [PMID: 39758992 PMCID: PMC11699617 DOI: 10.1016/j.isci.2024.111502] [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: 06/05/2024] [Revised: 10/20/2024] [Accepted: 11/26/2024] [Indexed: 01/07/2025] Open
Abstract
Transforming growth factor β (TGF-β) is abundantly present in the tumor microenvironment, contributing to cancer progression. However, the regulatory mechanism by which TGF-β affects vascular endothelial cells (ECs) in the tumor microenvironment is not well understood. Herein, we generated tamoxifen-inducible TGF-β type II receptor (TβRII) knockout mice, specifically targeting ECs (TβRIIiΔEC), by crossbreeding TβRII-floxed mice with Pdgfb-icreER mice. We established tumor-bearing mice by transplanting Lewis lung carcinoma (LLC) cells. TβRIIiΔEC mice exhibited increased tumor angiogenesis with fragile new blood vessels, increased bleeding, and hypoxia compared to control mice. Consequently, the compromised tumor microenvironment precipitated a notable surge in circulating tumor cells. Paradoxically, lung metastasis showed a significant decline. This intriguing discrepancy was explained by a reduction in the engraftment between cancer cells and ECs. Disruption of TGF-β signaling downregulated CD44 on ECs, hindering cancer cell adhesion. These findings highlight TGF-β's role in promoting metastasis by modulating EC function.
Collapse
Affiliation(s)
- Kako Hanada
- Laboratory of Stem Cell Regulation, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yuki Saito
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Takahiro Takagi
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Mitsuki Go
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yota Nakano
- Laboratory of Biochemistry, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan
| | - Toshihiko Inagawa
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Hideyo Hirai
- Laboratory of Stem Cell Regulation, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Marcus Fruttiger
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Susumu Itoh
- Laboratory of Biochemistry, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan
| | - Fumiko Itoh
- Laboratory of Stem Cell Regulation, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
32
|
Liang C, Wang Y, Zhao R, Du J, Yao J, Khan AUR, Zhu Y, Xia H, Zhu T. Multifunctional hybrid poly(ester-urethane)urea/resveratrol electrospun nanofibers for a potential vascularizing matrix. SOFT MATTER 2024; 21:55-67. [PMID: 39624984 DOI: 10.1039/d4sm00937a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2024]
Abstract
The challenges for clinical application of small-diameter vascular graft are mainly acute/chronic thrombosis, inadequate endothelialization, intimal hyperplasia caused by inflammation, oxidative stress, and the mismatch of mechanical compliance after transplantation. How to construct an effective regenerative microenvironment through a material with uniform dispersion of active components is the premise of maintaining patency of a vascular graft. In this study, we have compounded poly(ester-urethane)urea (PEUU) with various optimized concentrations of resveratrol (Res) by homogeneous emulsion blending, followed by electrospinning into the hybrid PEUU/Res nanofibers (P/R-0, P/R-0.5, P/R-1.0, and P/R-1.5). Then the microstructure, surface wettability, mechanical properties, degradation, Res sustained release properties, hemocompatibility, and cytocompatibility of P/R were evaluated comprehensively. The results indicate that Res can be gradually released from the P/R, and both the hydrophilicity and antioxidant ability of the nanofiber gradually increase with the increase of Res content. Moreover, with the increase of Res, the viability and proliferation behavior of HUVECs were significantly improved. Meanwhile, tube formation and migration experiments showed that Res promoted the formation of a neovascularization network. In brief, it is concluded that P/R-1.0 is the optimal candidate with a uniform microstructure, moderate wettability, optimized mechanical properties, reliable hemocompatibility and cytocompatibility, and strongest ability to promote endothelial growth for the vascularizing matrix.
Collapse
Affiliation(s)
- Chen Liang
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, P. R. China.
| | - Yanan Wang
- Department of Minimally Invasive Spine Surgery, Shandong Wendeng Orthopedic Hospital, 1 Fengshan Rd., Weihai 264400, Shandong, P. R. China
| | - Renliang Zhao
- Orthopedics Research Institute, Trauma Medical Center, Department of Orthopedics, West China Hospital, Sichuan University, 37 Guoxue Ln., Chengdu 610041, Sichuan, P. R. China
| | - Juan Du
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, P. R. China.
| | - Jin Yao
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, P. R. China.
| | - Atta Ur Rehman Khan
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, P. R. China.
| | - Youwei Zhu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd., Shanghai 200025, P. R. China.
- Shanghai Key Laboratory of Pancreatic Neoplasms Translational Medicine, 197 Ruijin 2nd Rd., Shanghai 200025, P. R. China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd., Shanghai 200025, P. R. China
| | - Huitang Xia
- Department of Plastic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, 16766 Jingshi Rd., Jinan 250014, Shandong, P. R. China.
- Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, 16766 Jingshi Rd., Jinan 250014, Shangdong, P. R. China
| | - Tonghe Zhu
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, P. R. China.
| |
Collapse
|
33
|
Huang Y, Zhang C, Zhang L, Chen X, Fan W. Chemical Synthesis and Multihybridization of Small-Sized Hollow Mesoporous Organosilica Nanoparticles Toward Advanced Theranostics. Acc Chem Res 2024; 57:3465-3477. [PMID: 39576957 DOI: 10.1021/acs.accounts.4c00502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
As one of the most widely used nanomaterials, mesoporous silica nanoparticles (MSNs) have received extensive attraction due to their desirable physicochemical performances of high stability, large surface area, and tunable pore sizes. Besides, the U.S. Food and Drug Administration (FDA) has recognized that silica-based nanoparticles are generally safe for biomedical applications. However, the poor biodegradation and inert Si-O-Si framework of inorganic MSNs severely impair their diverse biomedical applications. A promising strategy to improve the physicochemical properties of MSNs is the incorporation of functional organic moieties into their framework to construct mesoporous organosilica nanoparticles (MONs), which exhibit distinct advantages over traditional inorganic MSNs, such as adjustable organosilica framework, excellent biocompatibility, stimuli-responsive biodegradability, and even improved therapeutic effects. Moreover, the emerging hollow-structured MONs (HMONs) with an internal cavity can offer a large drug loading capacity and thus become increasingly attractive and promising theranostic nanoplatforms in biomedicine. In recent years, numerous studies have delved into establishing multifunctional HMONs with sizes ranging in diameters from 50 to 200 nm for desirable biological responses. With the gradual deepening of research, small-sized HMONs with diameters below 50 nm (sub-50 nm HMONs) demonstrate unparalleled advantages in extending blood circulation time, reducing the risk of vascular occlusion, and achieving high tumor accumulation, thus leading to a growing interest in the design, development, and translation of sub-50 nm HMONs. However, the mechanism of the chemical synthesis and structural regulation of sub-50 nm HMONs is still unclear, which is detrimental to further structural hybridization and surface functionalization. In this account, we will focus on the structural design, chemical synthesis, adjustable framework hybridization, multifunctional surface modification, and versatile biomedical applications of small-sized HMONs. First, we will illustrate the chemical approaches for controllable synthesis of HMONs and the underlying mechanism of particle size regulation below 50 nm. Subsequently, the basic principles and design strategies of multihybridization of sub-50 nm HMONs based on framework hybridization, surface modulation, and in situ polymerization will be systematically discussed. Through diverse functionalization strategies, a series of sub-50 nm multihybridized HMONs-based nanotheranostics are established, and their applications in multimodal biomedical imaging and highly efficient synergistic treatment of various diseases (e.g., cancer, glaucoma, bacterial infection, etc.) will be accounted. Finally, we will summarize the current status and potential challenges of HMONs in clinic trials, as well as provide a comprehensive outlook on the future development of sub-50 nm HMONs. These innovative sub-50 nm HMONs hold the potential to introduce novel theranostic modalities for a variety of systemic disorders and to advance smart promising nanomedicine in the near future.
Collapse
Affiliation(s)
- Yuhang Huang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Cheng Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Liyuan Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Lower Kent Ridge Road, 4 Science Drive 2, Singapore 117544, Singapore
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
34
|
Kurebayashi Y, Sugimoto K, Tsujikawa H, Matsuda K, Nomura R, Ueno A, Masugi Y, Yamazaki K, Effendi K, Takeuchi H, Itoi T, Hasegawa Y, Abe Y, Kitago M, Ojima H, Sakamoto M. Spatial Dynamics of T- and B-Cell Responses Predicts Clinical Outcome of Resectable and Unresectable Hepatocellular Carcinoma. Clin Cancer Res 2024; 30:5666-5680. [PMID: 39417698 DOI: 10.1158/1078-0432.ccr-24-0479] [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: 02/12/2024] [Revised: 07/16/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE Immunotherapies have led to a paradigm shift in the treatment of hepatocellular carcinoma (HCC). Studies have revealed the single-cell catalogs of tumor-infiltrating immune cells and the trajectories of their differentiation. Nevertheless, the spatial distribution of these immune cells with distinct phenotypes in the tumor microenvironment and their clinicopathologic significance in resectable and unresectable HCCs are still largely unclear. EXPERIMENTAL DESIGN We analyzed the spatial dynamics of intratumoral CD4 and CD8 T cells and their association with B and plasma cells using 283 surgically resected HCC samples, 58 unresectable HCC samples before combined immunotherapy [atezolizumab plus bevacizumab (Atezo + Bev)], and autopsy specimens from 50 cases of advanced-stage HCC through multiplex IHC combined with transcriptomic and driver gene mutation analyses. Classification based on the spatial dynamics of T- and B-cell responses (refined immunosubtype) was developed, and its clinicopathologic significance was analyzed. RESULTS We found that stem-like CD4 and CD8 T cells were mainly observed in T-cell aggregates and T-cell zone of tertiary lymphoid structure (TLS). The differentiation of T follicular helper cells was associated with the development of TLS, whereas the differentiation of CXCL13-expressing CD4 TCXCL13 cells with a phenotype resembling T peripheral helper cells was associated with the development of the lymphoplasmacytic microenvironment. The refined immunosubtype could predict clinical outcomes of resectable HCC after surgery and unresectable HCC after Atezo + Bev therapy. The immune microenvironment of metastatic lesions tended to reflect those of primary lesions. CONCLUSIONS We revealed the spatial dynamics of T- and B-cell responses in HCC, which is closely associated with the clinical outcome after surgical resection or Atezo + Bev therapy.
Collapse
Affiliation(s)
- Yutaka Kurebayashi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Katsutoshi Sugimoto
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo, Japan
| | - Hanako Tsujikawa
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
- Department of Diagnostic Pathology, National Hospital Organization Saitama Hospital, Saitama, Japan
| | - Kosuke Matsuda
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Rui Nomura
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Akihisa Ueno
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
- Department of Diagnostic Pathology, Keio University Hospital, Tokyo, Japan
| | - Yohei Masugi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
- Department of Diagnostic Pathology, Keio University Hospital, Tokyo, Japan
| | - Ken Yamazaki
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
- Division of Molecular Pathology, Research Institute, Tochigi Cancer Center, Tochigi, Japan
| | - Kathryn Effendi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Hirohito Takeuchi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo, Japan
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo, Japan
| | - Yasushi Hasegawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuta Abe
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hidenori Ojima
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
- Division of Molecular Pathology, Research Institute, Tochigi Cancer Center, Tochigi, Japan
| | - Michiie Sakamoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
- School of Medicine, International University of Health and Welfare, Narita, Japan
| |
Collapse
|
35
|
Cui JY, Ma J, Gao XX, Sheng ZM, Pan ZX, Shi LH, Zhang BG. Unraveling the role of cancer-associated fibroblasts in colorectal cancer. World J Gastrointest Oncol 2024; 16:4565-4578. [PMID: 39678792 PMCID: PMC11577382 DOI: 10.4251/wjgo.v16.i12.4565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 11/12/2024] Open
Abstract
Within the intricate milieu of colorectal cancer (CRC) tissues, cancer-associated fibroblasts (CAFs) act as pivotal orchestrators, wielding considerable influence over tumor progression. This review endeavors to dissect the multifaceted functions of CAFs within the realm of CRC, thereby highlighting their indispensability in fostering CRC malignant microenvironment and indicating the development of CAFs-targeted therapeutic interventions. Through a comprehensive synthesis of current knowledge, this review delineates insights into CAFs-mediated modulation of cancer cell proliferation, invasiveness, immune evasion, and neovascularization, elucidating the intricate web of interactions that sustain the pro-tumor metabolism and secretion of multiple factors. Additionally, recognizing the high level of heterogeneity within CAFs is crucial, as they encompass a range of subtypes, including myofibroblastic CAFs, inflammatory CAFs, antigen-presenting CAFs, and vessel-associated CAFs. Innovatively, the symbiotic relationship between CAFs and the intestinal microbiota is explored, shedding light on a novel dimension of CRC pathogenesis. Despite remarkable progress, the orchestrated dynamic functions of CAFs remain incompletely deciphered, underscoring the need for continued research endeavors for therapeutic advancements in CRC management.
Collapse
Affiliation(s)
- Jia-Yu Cui
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Jing Ma
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Xin-Xin Gao
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Zhi-Mei Sheng
- Affiliated Hospital of Shandong Second Medical University, Department of Pathology, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Zi-Xin Pan
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Li-Hong Shi
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Bao-Gang Zhang
- Department of Pathology, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| |
Collapse
|
36
|
Wang L, Ge Y, Yin L, Zong D, Li Y, Wu J, He X. Dynamic changes in serum adenosine and the adenosine metabolism-based signature for prognosis in HER2-positive metastatic breast cancer patients. Heliyon 2024; 10:e39545. [PMID: 39669152 PMCID: PMC11636132 DOI: 10.1016/j.heliyon.2024.e39545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 09/26/2024] [Accepted: 10/16/2024] [Indexed: 12/14/2024] Open
Abstract
Aims Adenosine metabolism in the breast cancer microenvironment is critical for tumor immunity. However, the prognostic significance of adenosine in breast cancer remains unclear. We aimed to dynamically monitor serum adenosine levels in patients with HER2-positive metastatic breast cancer (MBC) patients and to explore its predictive significance in trastuzumab therapy. Methods The sequencing and clinical data were downloaded from TCGA and GSE176078. Adenosine-related differentially expressed genes was analyzed by "DESeq2" package. Multivariate Cox and lasso-penalized Cox regressions were used to construct prognostic risk signatures. The risk scores were calculated from the identified expression of the hub genes. Bioinformatic analyses were performed using R with related packages. We also enrolled the metastatic breast cancer patients with HER2-positive from in our center and classified them into different groups according to the clinical outcomes assessed by enhanced CT. The adenosine levels were dynamically detected, and the difference in immune microenvironment between the subgroups was assessed by the immune cells that were recorded in our center. Results A total of 109 breast cancer patients with HER2-positive MBC were enrolled, and the expressions of 22 adenosine-related genes were filtered and matched from the TCGA database. The survival model based on the 15 differentially expressed genes was established, and the risk scores of each patient were the prognostic risk factors. Single-cell transcriptome sequencing data identified transcriptomic differences in patients with HER2-positive breast cancer. We also confirmed the predictive value of serum adenosine in the clinical progression of HER2-positive MBC patients. The different immune microenvironment between the subgroups supported the reliability of the predictive ability of adenosine in HER2-positive MBC patients. Conclusions The dynamic change of adenosine is a predictive biomarker for monitoring disease progression. The adenosine metabolism-based signature has the potential application in the prognosis of HER2-positive MBC patients.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Yizhi Ge
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Li Yin
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Dan Zong
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Yang Li
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Jianfeng Wu
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Xia He
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| |
Collapse
|
37
|
Youssef E, Zhao S, Purcell C, Olson GL, El-Deiry WS. Targeting the SMURF2-HIF1α axis: a new frontier in cancer therapy. Front Oncol 2024; 14:1484515. [PMID: 39697237 PMCID: PMC11652374 DOI: 10.3389/fonc.2024.1484515] [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: 08/22/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024] Open
Abstract
The SMAD-specific E3 ubiquitin protein ligase 2 (SMURF2) has emerged as a critical regulator in cancer biology, modulating the stability of Hypoxia-Inducible Factor 1-alpha (HIF1α) and influencing a network of hypoxia-driven pathways within the tumor microenvironment (TME). SMURF2 targets HIF1α for ubiquitination and subsequent proteasomal degradation, disrupting hypoxic responses that promote cancer cell survival, metabolic reprogramming, angiogenesis, and resistance to therapy. Beyond its role in HIF1α regulation, SMURF2 exerts extensive control over cellular processes central to tumor progression, including chromatin remodeling, DNA damage repair, ferroptosis, and cellular stress responses. Notably, SMURF2's ability to promote ferroptotic cell death through GSTP1 degradation offers an alternative pathway to overcome apoptosis resistance, expanding therapeutic options for refractory cancers. This review delves into the multifaceted interactions between SMURF2 and HIF1α, emphasizing how their interplay impacts metabolic adaptations like the Warburg effect, immune evasion, and therapeutic resistance. We discuss SMURF2's dual functionality as both a tumor suppressor and, in certain contexts, an oncogenic factor, underscoring its potential as a highly versatile therapeutic target. Furthermore, modulating the SMURF2-HIF1α axis presents an innovative approach to destabilize hypoxia-dependent pathways, sensitizing tumors to chemotherapy, radiotherapy, and immune-based treatments. However, the complexity of SMURF2's interactions necessitate a thorough assessment of potential off-target effects and challenges in specificity, which must be addressed to optimize its clinical application. This review concludes by proposing future directions for research into the SMURF2-HIF1α pathway, aiming to refine targeted strategies that exploit this axis and address the adaptive mechanisms of aggressive tumors, ultimately advancing the landscape of precision oncology.
Collapse
Affiliation(s)
- Emile Youssef
- Research & Development, SMURF-Therapeutics, Inc., Providence, RI, United States
- Medical & Pharmacovigilance, Kapadi, Inc., Raleigh, NC, United States
| | - Shuai Zhao
- Department of Pathology & Laboratory Medicine, Legorreta Cancer Center at Brown University, Providence, RI, United States
| | - Connor Purcell
- Department of Pathology & Laboratory Medicine, Legorreta Cancer Center at Brown University, Providence, RI, United States
| | - Gary L. Olson
- Medicinal Chemistry & Drug Discovery, Provid Pharmaceuticals, Inc., Monmouth Junction, NJ, United States
| | - Wafik S. El-Deiry
- Research & Development, SMURF-Therapeutics, Inc., Providence, RI, United States
- Department of Pathology & Laboratory Medicine, Legorreta Cancer Center at Brown University, Providence, RI, United States
| |
Collapse
|
38
|
Santio NM, Ganesh K, Kaipainen PP, Halme A, Seyednasrollah F, Arbash E, Hänninen S, Kivelä R, Carpen O, Saharinen P. Endothelial Pim3 kinase protects the vascular barrier during lung metastasis. Nat Commun 2024; 15:10514. [PMID: 39627185 PMCID: PMC11615401 DOI: 10.1038/s41467-024-54445-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/07/2024] [Indexed: 12/06/2024] Open
Abstract
Endothelial cells (ECs) form a tissue-specific barrier for disseminating cancer cells in distant organs. However, the molecular regulation of the ECs in the metastatic niche remains unclear. Here, we analyze using scRNA-Seq, the transcriptional reprogramming of lung ECs six hours after the arrival of melanoma cells in mouse lungs. We discover a reactive capillary EC cluster (rCap) that increases from general capillary ECs in response to infiltrating cancer cells. rCap is enriched for angiogenic and inflammatory pathways and is also found in human lung datasets. The JAK-STAT activated oncogenic Pim3 kinase is a marker of rCap, being upregulated in spontaneous metastasis models. Notably, PIM inhibition increases vascular leakage and metastatic colonization and impairs the EC barrier by decreasing the junctional cadherin-5 and catenins α, β and δ. These results highlight the pulmonary endothelium's plasticity and its protection by PIM3, which may impair the efficacy of PIM inhibitors in cancer therapies.
Collapse
Affiliation(s)
- Niina M Santio
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Keerthana Ganesh
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Pihla P Kaipainen
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Aleksi Halme
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Fatemeh Seyednasrollah
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Emad Arbash
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Satu Hänninen
- Systems Oncology, Research Programs Unit University of Helsinki, Finland, Helsinki
| | - Riikka Kivelä
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland
- Faculty of Sport and Health Sciences University of Jyväskylä, Jyväskylä, Finland
| | - Olli Carpen
- Systems Oncology, Research Programs Unit University of Helsinki, Finland, Helsinki
- Pathology/HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Pipsa Saharinen
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland.
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland.
- Department of Biochemistry and Developmental Biology, Faculty of Medicine University of Helsinki, Helsinki, Finland.
| |
Collapse
|
39
|
Xu Q, Shao D. Leveraging the synergy between anti-angiogenic therapy and immune checkpoint inhibitors to treat digestive system cancers. Front Immunol 2024; 15:1487610. [PMID: 39691707 PMCID: PMC11649667 DOI: 10.3389/fimmu.2024.1487610] [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: 08/28/2024] [Accepted: 11/20/2024] [Indexed: 12/19/2024] Open
Abstract
The response rates to immunotherapy vary widely depending on the type of cancer and the specific treatment used and can be disappointingly low for many solid tumors. Fortunately, due to their complementary mechanisms of action, immunotherapy and anti-angiogenic therapy have synergistic effects in cancer treatment. By normalizing the tumor vasculature, anti-angiogenic therapy can improve blood flow and oxygenation to facilitate better immune cell infiltration into the tumor and enhance the effectiveness of immunotherapy. It also reduces immunosuppressive factors and enhances immune activation, to create a more favorable environment for immune cells to attack the tumor. Their combination leverages the strengths of both therapies to enhance anti-tumor effects and improve patient outcomes. This review discusses the vasculature-immunity crosstalk in the tumor microenvironment and summarizes the latest advances in combining anti-angiogenic therapy and immune checkpoint inhibitors to treat digestive system tumors.
Collapse
Affiliation(s)
| | - Dong Shao
- Department of Gastroenterology, The Third Affiliated Hospital of Soochow
University, Changzhou, Jiangsu, China
| |
Collapse
|
40
|
Yang C, Liu W, Powell CA, Wang Q. Heterogeneity and therapeutic implications of cancer-associated fibroblasts in lung cancer: Recent advances and future perspectives. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:240-249. [PMID: 39834587 PMCID: PMC11742357 DOI: 10.1016/j.pccm.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Indexed: 01/22/2025]
Abstract
Lung cancer is a leading cause of cancer-related mortality. The tumor microenvironment is a complex and heterogeneous cellular environment surrounding tumor cells, including cancer-associated fibroblasts (CAFs), blood vessels, immune cells, the extracellular matrix, and various cytokines secreted by cells. CAFs are highly heterogeneous and play crucial roles in lung cancer. This review highlights recent advances in the understanding of CAFs in lung cancer, focusing on their heterogeneity and functions in tumorigenesis, progression, angiogenesis, invasion, metastasis, therapy resistance, tumor immune suppression, and targeted therapy responses. Additionally, we explore the underlying mechanisms and the potential of CAFs as a target in the development of innovative therapies for lung cancer.
Collapse
Affiliation(s)
- Chunhui Yang
- Department of Clinical Laboratory, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
| | - Wenwen Liu
- Translational Research Center for Lung Cancer, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
| | - Charles A. Powell
- Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Qi Wang
- Translational Research Center for Lung Cancer, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
| |
Collapse
|
41
|
Skurikhin EG, Ermakova NN, Zhukova MA, Pan ES, Zharkikh IL, Pan VY, Kubatiev AA, Morozov SG, Skurikhina VE, Minakova MY, Pershina OV, Dygai AM. Consequences of Reprogrammed CD8 + T-Cell Therapy for Lewis Lung Carcinoma Cells and Neovasculogenesis in C57BL/6 Mice. Bull Exp Biol Med 2024; 178:244-249. [PMID: 39762692 DOI: 10.1007/s10517-025-06315-z] [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: 07/23/2024] [Indexed: 01/15/2025]
Abstract
We studied the effect of reprogrammed CD8+ T cells (rT cells) from the bone marrow of intact mice on tumor cells and neovasculogenesis in mice with orthotopic Lewis lung carcinoma (LLC). Reprogramming of T cells was carried out using a MEK inhibitor and a PD-1 blocker; the targeting of rT cells to tumor cells was achieved by preincubation with LLC cell lysate. It was shown that the antitumor effect of rT cells was based on apoptosis of tumor cells. In addition, cell therapy reduced the number of endothelial cells (CD45-CD309+) and angiogenic cell precursors (CD45-CD117+CD309+), mesenchymal stem cells (CD45-CD31-CD34-CD44+), myeloid (CD45+CD34+CD31-) and non-myeloid (CD45+CD34-CD31-) fibrocytes, and leukocytes (CD45+) in the lungs and increased their number in the blood. Thus, rT cells impaired the recruitment of neovasculogenic cells to the lung. The antitumor effects of rT cells are superior to those of naive CD8+ T cells. The proposed reprogramming method can be useful in developing effective approaches to the therapy of lung cancer, as it allows obtaining cytotoxic rT cells capable of reducing the activity of neovasculogenesis.
Collapse
Affiliation(s)
- E G Skurikhin
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - N N Ermakova
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - M A Zhukova
- Institute of General Pathology and Pathophysiology, Moscow, Russia.
| | - E S Pan
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - I L Zharkikh
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - V Yu Pan
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - A A Kubatiev
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - S G Morozov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - V E Skurikhina
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - M Yu Minakova
- Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - O V Pershina
- Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - A M Dygai
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
42
|
Ielpo S, Barberini F, Dabbagh Moghaddam F, Pesce S, Cencioni C, Spallotta F, De Ninno A, Businaro L, Marcenaro E, Bei R, Cifaldi L, Barillari G, Melaiu O. Crosstalk and communication of cancer-associated fibroblasts with natural killer and dendritic cells: New frontiers and unveiled opportunities for cancer immunotherapy. Cancer Treat Rev 2024; 131:102843. [PMID: 39442289 DOI: 10.1016/j.ctrv.2024.102843] [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: 08/27/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
Natural killer (NK) cells and dendritic cells (DCs) are critical mediators of anti-cancer immune responses. In addition to their individual roles, NK cells and DCs are involved in intercellular crosstalk which is essential for the initiation and coordination of adaptive immunity against cancer. However, NK cell and DC activity is often compromised in the tumor microenvironment (TME). Recently, much attention has been paid to one of the major components of the TME, the cancer-associated fibroblasts (CAFs), which not only contribute to extracellular matrix (ECM) deposition and tumor progression but also suppress immune cell functions. It is now well established that CAFs support T cell exclusion from tumor nests and regulate their cytotoxic activity. In contrast, little is currently known about their interaction with NK cells, and DCs. In this review, we describe the interaction of CAFs with NK cells and DCs, by secreting and expressing various mediators in the TME of adult solid tumors. We also provide a detailed overview of ongoing clinical studies evaluating the targeting of stromal factors alone or in combination with immunotherapy based on immune checkpoint inhibitors. Finally, we discuss currently available strategies for the selective depletion of detrimental CAFs and for a better understanding of their interaction with NK cells and DCs.
Collapse
Affiliation(s)
- Simone Ielpo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Barberini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Farnaz Dabbagh Moghaddam
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, Rome, Italy
| | - Silvia Pesce
- Department of Experimental Medicine and Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Chiara Cencioni
- Institute for Systems Analysis and Computer Science "A. Ruberti", National Research Council (IASI-CNR), Rome, Italy
| | - Francesco Spallotta
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University, 00185, Rome, Italy; Pasteur Institute Italy-Fondazione Cenci Bolognetti, Italy
| | - Adele De Ninno
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, Rome, Italy
| | - Luca Businaro
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, Rome, Italy
| | - Emanuela Marcenaro
- Department of Experimental Medicine and Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Loredana Cifaldi
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
43
|
Tang L, He D, Su B. Nrf2: A critical participant in regulation of apoptosis, ferroptosis, and autophagy in gastric cancer. Acta Histochem 2024; 126:152203. [PMID: 39342913 DOI: 10.1016/j.acthis.2024.152203] [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: 05/05/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
Nuclear factor erythroid 2-related factor-2 (Nrf2) is a specific transcription factor that maintains redox homeostasis by regulating the expression of anti-oxidative stress-related genes. Hyperactivation of Nrf2 is involved in tumor progression and is associated with chemoresistance in a large number of solid tumors. Programmatic cell death (PCD), such as apoptosis, ferroptosis, and autophagy, plays a crucial role in tumor development and chemotherapy sensitivity. Accumulating evidence suggests that some anti-tumor compounds and genes can induce massive production of reactive oxygen species (ROS) via inhibiting Nrf2 expression, which exacerbates oxidative stress and promotes Gastric cancer (GC) cell death, thereby enhancing the sensitivity of GC cells to chemotherapy-induced PCD. In this review, we summarize the role of antitumor drugs in interfering in three different types of PCD (apoptosis, ferroptosis, and autophagy) in GC cells by modulating Nrf2 expression, as well as the molecular mechanisms through which targeting Nrf2 brings about PCD and chemosensitivity. It is reasonable to believe that Nrf2 serves as a potential therapeutic target, and targeting Nrf2 by drug or gene regulation could provide a new strategy for the treatment of GC.
Collapse
Affiliation(s)
- LiJie Tang
- Institute of Pharmacy and Pharmacology, School of Pharmacy, Hengyang Medical School, University of South China, Hengyang, China
| | - DongXiu He
- Institute of Pharmacy and Pharmacology, School of Pharmacy, Hengyang Medical School, University of South China, Hengyang, China
| | - Bo Su
- Institute of Pharmacy and Pharmacology, School of Pharmacy, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
44
|
Wu J, Chen Y, Xie M, Yu X, Su C. cGAS-STING signaling pathway in lung cancer: Regulation on antitumor immunity and application in immunotherapy. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:257-264. [PMID: 39834588 PMCID: PMC11742360 DOI: 10.1016/j.pccm.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 08/31/2024] [Accepted: 11/03/2024] [Indexed: 01/22/2025]
Abstract
The innate immune system has a primary role in defending against external threats, encompassing viruses, bacteria, and fungi, thereby playing a pivotal role in establishing robust protection. Recent investigations have shed light on its importance in the progression of tumors, with a particular emphasis on lung cancer. Among the various signaling pathways implicated in this intricate process, the cGAS-STING pathway emerges as a significant participant. Cyclic GMP-AMP synthase (cGAS) discerns free DNA and activates the stimulator of interferon genes (STING), subsequently culminating in the secretion of cytokines and exerting inhibitory effects on tumor development. Consequently, researchers are increasingly interested in creating anticancer drugs that specifically target the cGAS-STING pathway, offering promising avenues for novel therapeutic interventions. The objective of this review is to present a comprehensive overview of the ongoing research on the cGAS-STING signaling pathway within the realm of lung cancer. The primary emphasis is on understanding its involvement in lung cancer development and assessing its viability as a target for innovative therapeutic options.
Collapse
Affiliation(s)
- Jing Wu
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Yingyao Chen
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Mengqing Xie
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Xin Yu
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Chunxia Su
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
45
|
Moynihan A, Boland P, Cucek J, Erzen S, Hardy N, McEntee P, Rojc J, Cahill R. Technical and functional design considerations for a real-world interpretable AI solution for NIR perfusion analysis (including cancer). EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108273. [PMID: 38538505 DOI: 10.1016/j.ejso.2024.108273] [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: 11/29/2023] [Revised: 03/03/2024] [Accepted: 03/16/2024] [Indexed: 12/02/2024]
Abstract
Near infrared (NIR) analysis of tissue perfusion via indocyanine green fluorescence assessment is performed clinically during surgery for a range of indications. Its usefulness can potentially be further enhanced through the application of interpretable artificial intelligence (AI) methods to improve dynamic interpretation accuracy in these and also open new applications. While its main use currently is for perfusion assessment as a tissue health check prior to performing an anastomosis, there is increasing interest in using fluorophores for cancer detection during surgical interventions with most research being based on the paradigm of static imaging for fluorophore uptake hours after preoperative dosing. Although some image boosting and relative estimation of fluorescence signals is already inbuilt into commercial NIR systems, fuller implementation of AI methods can enable actionable predictions especially when applied during the dynamic, early inflow-outflow phase that occurs seconds to minutes after ICG (or indeed other fluorophore) administration. Already research has shown that such methods can accurately differentiate cancer from benign tissue in the operating theatre in real time in principle based on their differential signalling and could be useful for tissue perfusion classification more generally. This can be achieved through the generation of fluorescence intensity curves from an intra-operative NIR video stream. These curves are processed to adjust for image disturbances and curve features known to be influential in tissue characterisation are extracted. Existing machine learning based classifiers can then use these features to classify the tissue in question according to prior training sets. The use of this interpretable methodology enables accurate classification algorithms to be built with modest training sets in comparison to those required for deep learning modelling in addition to achieving compliance with medical device regulations. Integration of the multiple algorithms required to achieve this classification into a desktop application or medical device could make the use of this method accessible and useful to (as well as useable by) surgeons without prior training in computer technology. This document details some technical and functional design considerations underlying such a novel recommender system to advance the foundational concept and methodology as software as medical device for in situ cancer characterisation with relevance more broadly also to other tissue perfusion applications.
Collapse
Affiliation(s)
- A Moynihan
- UCD Centre for Precision Surgery, University College Dublin, Ireland
| | - P Boland
- UCD Centre for Precision Surgery, University College Dublin, Ireland
| | - J Cucek
- Arctur, Nova Gorica, Slovenia
| | - S Erzen
- Arctur, Nova Gorica, Slovenia
| | - N Hardy
- UCD Centre for Precision Surgery, University College Dublin, Ireland
| | - P McEntee
- UCD Centre for Precision Surgery, University College Dublin, Ireland
| | - J Rojc
- Arctur, Nova Gorica, Slovenia
| | - R Cahill
- UCD Centre for Precision Surgery, University College Dublin, Ireland; Department of Surgery, Mater Misericordiae University Hospital, Dublin, Ireland.
| |
Collapse
|
46
|
Surya C, Lakshminarayana ABV, Ramesh SH, Kunjiappan S, Theivendren P, Santhana Krishna Kumar A, Ammunje DN, Pavadai P. Advancements in breast cancer therapy: The promise of copper nanoparticles. J Trace Elem Med Biol 2024; 86:127526. [PMID: 39298835 DOI: 10.1016/j.jtemb.2024.127526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/12/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Breast cancer (BC) is the most prevalent cancer among women worldwide and poses significant treatment challenges. Traditional therapies often lead to adverse side effects and resistance, necessitating innovative approaches for effective management. OBJECTIVE This review aims to explore the potential of copper nanoparticles (CuNPs) in enhancing breast cancer therapy through targeted drug delivery, improved imaging, and their antiangiogenic properties. METHODS The review synthesizes existing literature on the efficacy of CuNPs in breast cancer treatment, addressing common challenges in nanotechnology, such as nanoparticle toxicity, scalability, and regulatory hurdles. It proposes a novel hybrid method that combines CuNPs with existing therapeutic modalities to optimize treatment outcomes. RESULTS CuNPs demonstrate the ability to selectively target cancer cells while sparing healthy tissues, leading to improved therapeutic efficacy. Their unique physicochemical properties facilitate efficient biodistribution and enhanced imaging capabilities. Additionally, CuNPs exhibit antiangiogenic activity, which can inhibit tumor growth by preventing the formation of new blood vessels. CONCLUSION The findings suggest that CuNPs represent a promising avenue for advancing breast cancer treatment. By addressing the limitations of current therapies and proposing innovative solutions, this review contributes valuable insights into the future of nanotechnology in oncology.
Collapse
Affiliation(s)
- Chandana Surya
- Department of Pharmacognosy, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka 560054, India
| | | | - Sameera Hammigi Ramesh
- Department of Pharmacology, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka 560054, India
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Tamilnadu 626126, India
| | - Panneerselvam Theivendren
- Department of Pharmaceutical Chemistry, Swamy Vivekananda College of Pharmacy, Elayampalayam, Namakkal, Tamilnadu 637205, India
| | - A Santhana Krishna Kumar
- Department of Chemistry, National Sun Yat-sen University, No. 70, Lien-hai Road, Gushan District, Kaohsiung City 80424, Taiwan; Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu 602105, India.
| | - Damodar Nayak Ammunje
- Department of Pharmacology, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka 560054, India.
| | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka 560054, India.
| |
Collapse
|
47
|
Maggi E, Munari E, Landolina N, Mariotti FR, Azzarone B, Moretta L. T cell landscape in the microenvironment of human solid tumors. Immunol Lett 2024; 270:106942. [PMID: 39486594 DOI: 10.1016/j.imlet.2024.106942] [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: 10/03/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
T cells are the main effectors involved in anti-tumor immunity, mediating most of the adaptive response towards cancer. After priming in lymph nodes, tumor antigens-specific naïve T lymphocytes proliferate and differentiate into effector CD4+ and CD8+ T cells that migrate from periphery into tumor sites aiming to eliminate cancer cells. Then while most effector T cells die, a small fraction persists and recirculates as long-lived memory T cells which generate enhanced immune responses when re-encountering the same antigen. A number of T (and non-T) cell subsets, stably resides in non-lymphoid peripheral tissues and may provide rapid immune response independently of T cells recruited from blood, against the reemergence of cancer cells. When tumor grows, however, tumor cells have evaded immune surveillance of effector cells (NK and CTL cells) which are exhausted, thus favoring the local expansion of T (and non-T) regulatory cells. In this review, the current knowledge of features of T cells present in the tumor microenvironment (TME) of solid adult and pediatric tumors, the mechanisms upregulating immune-checkpoint molecules and transcriptional and epigenetic landscapes leading to dysfunction and exhaustion of T effector cells are reviewed. The interaction of T cells with cancer- or TME non-neoplastic cells and their secreted molecules shape the T cell profile compromising the intrinsic plasticity of T cells and, therefore, favoring immune evasion. In this phase regulatory T cells contribute to maintain a high immunosuppressive TME thus facilitating tumor cell proliferation and metastatic spread. Despite the advancements of cancer immunotherapy, many tumors are unresponsive to immune checkpoint inhibitors, or therapeutical vaccines or CAR T cell-based adoptive therapy: some novel strategies to improve these T cell-based treatments are lastly proposed.
Collapse
Affiliation(s)
- Enrico Maggi
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | - Enrico Munari
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona 37126, Italy
| | - Nadine Landolina
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | | | - Bruno Azzarone
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy.
| |
Collapse
|
48
|
Urra H, Aravena R, González-Johnson L, Hetz C. The UPRising connection between endoplasmic reticulum stress and the tumor microenvironment. Trends Cancer 2024; 10:1161-1173. [PMID: 39472237 DOI: 10.1016/j.trecan.2024.09.011] [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: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 12/12/2024]
Abstract
The tumor microenvironment (TME) represents a dynamic network of cancer cells, stromal cells, immune mediators, and extracellular matrix components, crucial for cancer progression. Stress conditions such as oncogene activation, nutrient deprivation, and hypoxia disrupt the endoplasmic reticulum (ER), activating the unfolded protein response (UPR), the main adaptive mechanism to restore ER function. The UPR regulates cancer progression by engaging cell-autonomous and cell-non-autonomous mechanisms, reprogramming the stroma and promoting immune evasion, angiogenesis, and invasion. This review explores the role of UPR beyond cancer cells, focusing on how ER stress signaling reshapes the TME, supporting tumor growth. The therapeutic potential of targeting the UPR is also discussed.
Collapse
Affiliation(s)
- Hery Urra
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile; Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Raúl Aravena
- Centro de Biología Celular y Biomedicina, Facultad de Medicina y Ciencia (CEBICEM), Universidad San Sebastián, Santiago 7510602, Chile
| | - Lucas González-Johnson
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile; Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; Department of Neurology and Neurosurgery, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Claudio Hetz
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile; Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile; The Buck Institute for Research in Aging, Novato, CA, USA.
| |
Collapse
|
49
|
Luan Y, Jia R, Chai P, Fan X. m 6A and beyond: RNA modifications shaping angiogenesis. Trends Mol Med 2024:S1471-4914(24)00302-2. [PMID: 39609142 DOI: 10.1016/j.molmed.2024.11.001] [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: 08/28/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024]
Abstract
RNA modifications are crucial post-transcriptional processes that significantly influence gene expression, RNA stability, nuclear transport, and translational capacity. Angiogenesis, the formation of new blood vessels, is a physiological process that is dysregulated in many pathological conditions, including ocular diseases, immune disorders, and cancer. In this review, we compile the current understanding of the intricate relationship between various RNA modifications and angiogenic mechanisms, spotlighting emerging evidence that underscore their pivotal regulatory roles in both physiological and pathological angiogenesis. Furthermore, we delve into recent advances in innovative therapeutic approaches that target RNA modifications to modulate angiogenesis, offering insights into their potential as novel treatment modalities.
Collapse
Affiliation(s)
- Yu Luan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| |
Collapse
|
50
|
Weerasinghe HN, Burrage PM, Jr DVN, Burrage K. Agent-based modeling for the tumor microenvironment (TME). MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:7621-7647. [PMID: 39696854 DOI: 10.3934/mbe.2024335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Cancer is a disease that arises from the uncontrolled growth of abnormal (tumor) cells in an organ and their subsequent spread into other parts of the body. If tumor cells spread to surrounding tissues or other organs, then the disease is life-threatening due to limited treatment options. This work applies an agent-based model to investigate the effect of intra-tumoral communication on tumor progression, plasticity, and invasion, with results suggesting that cell-cell and cell-extracellular matrix (ECM) interactions affect tumor cell behavior. Additionally, the model suggests that low initial healthy cell densities and ECM protein densities promote tumor progression, cell motility, and invasion. Furthermore, high ECM breakdown probabilities of tumor cells promote tumor invasion. Understanding the intra-tumoral communication under cellular stress can potentially lead to the design of successful treatment strategies for cancer.
Collapse
Affiliation(s)
- Hasitha N Weerasinghe
- School of Mathematical Sciences, Queensland University of Technology, Queensland, Brisbane, Australia
| | - Pamela M Burrage
- School of Mathematical Sciences, Queensland University of Technology, Queensland, Brisbane, Australia
| | - Dan V Nicolau Jr
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Kevin Burrage
- School of Mathematical Sciences, Queensland University of Technology, Queensland, Brisbane, Australia
- Department of Computer Science, University of Oxford, United Kingdom
| |
Collapse
|