1
|
Oliveira Dias J, Sampaio Fagundes I, Bisio MDC, da Silva Barboza V, Jacinto AA, Altei WF. Extracellular vesicles as the common denominator among the 7 Rs of radiobiology: From the cellular level to clinical practice. Biochim Biophys Acta Rev Cancer 2025; 1880:189315. [PMID: 40216093 DOI: 10.1016/j.bbcan.2025.189315] [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: 11/22/2024] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Extracellular vesicles (EVs) are lipid-bound particles released by tumor cells and widely explored in cancer development, progression, and treatment response, being considered as valuable components to be explored as biomarkers or cellular targets to modulate the effect of therapies. The mechanisms underlying the production and profile of EVs during radiotherapy (RT) require addressing radiobiological aspects to determine cellular responses to specific radiation doses and fractionation. In this review, we explore the role of EVs in the 7 Rs of radiobiology, known as the molecular basis of a biological tissue response to radiation, supporting EVs as a shared player in all the seven processes. We also highlight the relevance of EVs in the context of liquid biopsy and resistance to immunotherapy, aiming to establish the connection and utility of EVs as tools in contemporary and precision radiotherapy.
Collapse
Affiliation(s)
- Júlia Oliveira Dias
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | | | | | - Wanessa Fernanda Altei
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil; Radiation Oncology Department, Barretos Cancer Hospital, Barretos, Brazil.
| |
Collapse
|
2
|
Swati, Basak P, Mittal BR, Shukla J, Chadha VD. Systemic effects of 177Lu-DOTATATE therapy to patients with metastatic neuroendocrine tumors: mechanistic insights and role of exosome. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07291-2. [PMID: 40263207 DOI: 10.1007/s00259-025-07291-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE The present study aimed to evaluate the systemic redox status in metastatic neuroendocrine tumor (NET) patients following 177Lu-DOTATATE therapy and to explore the role of exosomes in communicating the redox signals in-vitro. METHODS Levels of reactive oxygen species (ROS), enzymes associated with oxidative stress and lipid peroxidation, gene expression of oxidative stress markers (COX2, iNOS, NF-κB and SOD) were determined in peripheral blood mononuclear cells (PBMCs) and serum isolated from a total of 30 NET subjects at three time points viz.: before, 4 weeks after first and fourth cycle of 177Lu-DOTATATE therapy. Serum cytokine levels (IL-2, IL-6, IFN-γ, TNF-α, IL-4, IL-10 and TGF-β) were measured by ELISA. DNA damage was assessed by checking the expression of γH2AX and DNA repair genes (ATM: Ataxia-Telangiectasia Mutated and ATR: Ataxia-Telangiectasia and Rad3-related). Plasma-derived exosomes were characterized, their uptake by PBMCs was visualized and consequent ROS generation was assessed in in-vitro co-culture. RESULTS The study exhibits a significant increase in ROS level and relatively higher expression of COX2 and iNOS in PBMCs of NET patients post therapy. Serum inflammatory cytokines including IL-2, IL-6 and TNF-α were found elevated. The study did not find any change in the expression of genes associated with DNA damage. In-vitro co-culture of PBMCs (isolated before therapy) with exosomes derived after therapy exhibited significant increase in ROS as compared to control cells. CONCLUSION The study concludes that 177Lu-DOTATATE therapy alters redox status, however it does not cause DNA damage, suggestive of its safety. Further, the study demonstrates the role of exosomes in spreading of oxidative stress systemically.
Collapse
Affiliation(s)
- Swati
- Centre for Nuclear Medicine (UIEAST), Panjab University, Chandigarh, India
| | - Preetam Basak
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - B R Mittal
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Jaya Shukla
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | |
Collapse
|
3
|
S BR, Dhar R, Devi A. Exosomes-mediated CRISPR/Cas delivery: A cutting-edge frontier in cancer gene therapy. Gene 2025; 944:149296. [PMID: 39884405 DOI: 10.1016/j.gene.2025.149296] [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: 12/10/2024] [Revised: 01/09/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Cancer is considered the second most common disease globally. In the past few decades, many approaches have been proposed for cancer treatment. One among those is targeted therapy using CRISPR/Cas system which plays a significant role in translational research through gene editing. However, due to its inability to cope with specific targeting, off-target effects, and limited tumor penetration, it is very challenging to use this approach in cancer studies. To increase its efficacy, CRISPR components are engineered into the extracellular vesicles (EVs), especially exosomes (a subpopulation of EVs). Exosomes have a significant role in cellular communication. Exosomes-based CRISPR/Cas system transport for gene editing enhances specificity, reduces off-target effects, and improves the therapeutic potential. This review highlights the role of exosomes and the CRISPR/Cas system in cancer research, exosomes-based CRISPR delivery for cancer treatment, and its future orientation.
Collapse
Affiliation(s)
- Bhavanisha Rithiga S
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, TamilNadu 603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, TamilNadu 603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, TamilNadu 603203, India.
| |
Collapse
|
4
|
Terashima S, Tatemura R, Saito W, Hosokawa Y. Evaluation of the influence of radiation-induced cohort effect in cell populations receiving different doses. Int J Radiat Biol 2025; 101:341-350. [PMID: 39899278 DOI: 10.1080/09553002.2025.2459086] [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: 07/15/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE A non-targeted effect called radiation-induced cohort effect, which results in interactions among irradiated neighboring cells through cellular communication, has been reported. In high-precision radiotherapy, the dose is localized to the tumor, and rapid spatial changes occur in dose distribution. However, the effect of irradiating a population of cells with non-uniform doses remains unknown. In this study, we evaluated the influence of cohort effect by creating cell populations irradiated with different doses using human oral squamous cell carcinoma (SAS) and human lung (A549) cells. MATERIALS AND METHODS Cell populations irradiated with different doses were created in two ways: direct contact co-culture (DCC) using a cell tracer dye and indirect contact co-culture (ICC) using cell culture inserts to assess the effects of soluble factors. Target cells were irradiated with 4 Gy and co-cultured cells with 0, 0.8, 3.2, and 4 Gy. In DCC, cell proliferation assays were performed using a flow cytometer, and in ICC, modified high-density survival, clonogenic, and apoptosis assays were performed. RESULTS In DCC, irradiation of co-cultured cells with X-rays increased the relative proliferation rate of the target cells. Similarly, irradiating co-cultured cells using ICC with X-rays increased the relative survival rate of target cells. CONCLUSIONS The results of this study showed that, even if there is a sharp decrease in dose near the tumor, the cytocidal effect on the tumor is not adversely affected. In addition, soluble factors were found to be involved in cohort effect.
Collapse
Affiliation(s)
- Shingo Terashima
- Department of Radiation Science, Hirosaki University Graduate School of Health Sciences, Hirosaki, Japan
| | - Ryota Tatemura
- Department of Radiology, Division of Medical Technology, Hirosaki University School of Medicine and Hospital, Hirosaki, Japan
| | - Wataru Saito
- Plant Operation Department, Reprocessing Plant, Reprocessing Business Division, Japan Nuclear Fuel Limited, Rokkasho-mura, Japan
| | - Yoichiro Hosokawa
- Department of Rehabilitation Sciences, Hirosaki University of Health and Welfare, Hirosaki, Japan
| |
Collapse
|
5
|
Meena SS, Kosgei BK, Soko GF, Tingjun C, Chambuso R, Mwaiselage J, Han RPS. Developing anti-TDE vaccine for sensitizing cancer cells to treatment and metastasis control. NPJ Vaccines 2025; 10:18. [PMID: 39870669 PMCID: PMC11772600 DOI: 10.1038/s41541-024-01035-3] [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: 05/13/2024] [Accepted: 11/21/2024] [Indexed: 01/29/2025] Open
Abstract
Tumor-derived exosomes (TDEs) mediate oncogenic communication, which modifies target cells to reinforce a tumor-promoting microenvironment. TDEs support cancer progression by suppressing anti-tumor immune responses, promoting metastasis, and conferring drug resistance. Thus, targeting TDEs could improve the efficacy of anti-cancer treatments and control metastasis. Current strategies to inhibit TDE-mediated oncogenic communication including drug-based and genetic modification-based inhibition of TDE release and/or uptake, have proved to be inefficient. In this work, we propose TDE surface engineering to express foreign antigens that will trigger life-long anti-TDE immune responses. The possibility of combining the anti-TDE vaccines with other treatments such as chemotherapy, radiotherapy, targeted therapy, and surgery is also explored.
Collapse
Affiliation(s)
- Stephene S Meena
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
- Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
- Ocean Road Cancer Institute, Dar es Salaam, United Republic of Tanzania.
| | - Benson K Kosgei
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Geofrey F Soko
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Ocean Road Cancer Institute, Dar es Salaam, United Republic of Tanzania
| | - Cheng Tingjun
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Ramadhani Chambuso
- Department of Global Health and Population, Harvard Chan School of Public Health, Harvard University, Cambridge, MA, USA
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Julius Mwaiselage
- Ocean Road Cancer Institute, Dar es Salaam, United Republic of Tanzania
| | - Ray P S Han
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
- Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
| |
Collapse
|
6
|
Lach MS, Wróblewska JP, Michalak M, Budny B, Wrotkowska E, Suchorska WM. The Effect of Ionising Radiation on the Properties of Tumour-Derived Exosomes and Their Ability to Modify the Biology of Non-Irradiated Breast Cancer Cells-An In Vitro Study. Int J Mol Sci 2025; 26:376. [PMID: 39796230 PMCID: PMC11719956 DOI: 10.3390/ijms26010376] [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: 12/13/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
The vast majority of breast cancer patients require radiotherapy but some of them will develop local recurrences and potentially metastases in the future. Recent data show that exosomal cargo is essential in these processes. Thus, we investigated the influence of ionising radiation on exosome properties and their ability to modify the sensitivity and biology of non-irradiated cells. Exosomes were isolated from breast cancer cell lines (MDA-MB-231, MCF7, and SKBR3) irradiated with 2 Gy (Exo 2 Gy) or no irradiation (Exo 0 Gy). Despite some differences in their molecular profiles, they did not affect cell viability, proliferation, cell cycle phase distribution, and radioresistance; however, both populations showed the ability to modify cell migration and invasion potential, as confirmed by the downregulation of β-catenin, which is responsible for maintaining the epithelial phenotype. Interestingly, exosomes from irradiated BCa cells were more actively deposited in the endothelial cells (EA.hy926). Furthermore, exosomes tend to lower the expression of CD31, which is responsible for maintaining intact vascularity. This preliminary study demonstrates the vital role of exosomes and their altered profile due to irradiation in the pathobiology of breast cancer.
Collapse
Affiliation(s)
- Michał Stefan Lach
- Department of Electroradiology, Poznan University of Medical Sciences, Garbary 15, 61-866 Poznan, Poland;
- Radiobiology Lab, The Greater Poland Cancer Centre, Garbary 15 Street, 61-866 Poznan, Poland
| | - Joanna Patrycja Wróblewska
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 6, Avenue du Swing, 4367 Belvaux, Luxembourg;
| | - Marcin Michalak
- Surgical, Oncological and Endoscopic Gynaecology Department, The Greater Poland Cancer Centre, Garbary 15 Street, 61-866 Poznan, Poland;
| | - Bartłomiej Budny
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Przybyszewskiego 49 Street, 60-355 Poznan, Poland; (B.B.); (E.W.)
| | - Elżbieta Wrotkowska
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Przybyszewskiego 49 Street, 60-355 Poznan, Poland; (B.B.); (E.W.)
| | - Wiktoria Maria Suchorska
- Department of Electroradiology, Poznan University of Medical Sciences, Garbary 15, 61-866 Poznan, Poland;
- Radiobiology Lab, The Greater Poland Cancer Centre, Garbary 15 Street, 61-866 Poznan, Poland
| |
Collapse
|
7
|
Huang M, Ji J, Xu X, Jin D, Wu T, Lin R, Huang Y, Qian J, Tan Z, Jiang F, Hu X, Xu W, Xiao M. Known and unknown: Exosome secretion in tumor microenvironment needs more exploration. Genes Dis 2025; 12:101175. [PMID: 39524543 PMCID: PMC11550746 DOI: 10.1016/j.gendis.2023.101175] [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: 05/17/2023] [Revised: 09/06/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2024] Open
Abstract
Exosomes, extracellular vesicles originating from endosomes, were discovered in the late 1980s and their function in intercellular communication has since garnered considerable interest. Exosomes are lipid bilayer-coated vesicles that range in size from 30 to 150 nm and appear as sacs under the electron microscope. Exosome secretion is crucial for cell-to-cell contact in both normal physiology and the development and spread of tumors. Furthermore, cancer cells can secrete more exosomes than normal cells. Scientists believe that intercellular communication in the complex tissue environment of the human body is an important reason for cancer cell invasion and metastasis. For example, some particles containing regulatory molecules are secreted in the tumor microenvironment, including exosomes. Then the contents of exosomes can be released by donor cells into the environment and interact with recipient cells to promote the migration and invasion of tumor cells. Therefore, in this review, we summarized the biogenesis of exosome, as well as exosome cargo and related roles. More importantly, this review introduces and discusses the factors that have been reported to affect exosome secretion in tumors and highlights the important role of exosomes in tumors.
Collapse
Affiliation(s)
- Mengxiang Huang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Jie Ji
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xuebing Xu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Dandan Jin
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Tong Wu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Renjie Lin
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Yuxuan Huang
- Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Jiawen Qian
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Zhonghua Tan
- Department of Nuclear Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Feng Jiang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xiaogang Hu
- Department of Respiratory Medicine, Rudong County People's Hospital, Nantong, Jiangsu 226400, China
| | - Weisong Xu
- Department of Gastroenterology, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Mingbing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
8
|
Li Y, Long Y, Ge X, Zhang P, Li T, Wu L, Fan H, Du Z, Liu Q, Hu Y. Radiation-Induced Tumor-Derived Extracellular Vesicles Combined with Tyrosine Kinase Inhibitors: An Effective and Safe Therapeutic Approach for Lung Adenocarcinoma with EGFR19Del. Vaccines (Basel) 2024; 12:1412. [PMID: 39772073 PMCID: PMC11680254 DOI: 10.3390/vaccines12121412] [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: 10/22/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Combining radiotherapy with targeted therapy benefits patients with advanced epidermal growth factor receptor-mutated non-small cell lung cancer (EGFRm NSCLC). However, the optimal strategy to combine EGFR tyrosine kinase inhibitors (TKIs) with radiotherapy for maximum efficacy and minimal toxicity is still uncertain. Notably, EVs, which serve as communication mediators among tumor cells, play a crucial role in the anti-tumor immune response. Methods To exploit the role of EVs in the delivery of tumor antigens, we formulated a therapeutic strategy that involves the use of radiation-induced tumor-derived EVs (TEXs) loaded onto dendritic cells (DCs) as a kind of vaccine in conjunction with EGFR TKIs and assessed the efficacy and safety of this approach in the treatment of EGFRm NSCLC. Results In our study, we characterized the release of immunogens as influenced by various modes of cell death, examining the impact of different levels of cell death under diverse irradiation modalities. Our results demonstrated that a radiation mode of 6Gy*3f exhibited the most promising potential to stimulate anti-tumor immune responses. This radiotherapy fraction, combined with TKIs, showed promising results in a tumor-bearing mouse model with an EGFR mutation, although there is a risk of radiation-associated pneumonitis. Furthermore, we found that 6Gy*3f-TEXs in vitro activate DCs and promote T cell proliferation as well as cytotoxic T lymphocyte-mediated tumor cell destruction. The administration of EGFR-TKIs combined DCs loaded with 6Gy*3f-TEXs exhibited the potential to inhibit tumor growth and mitigate the risk of pneumonitis. Together, the research shows that TEXs from high-dose fractionation radiation can mature DCs and boost the killing of cytotoxic T lymphocytes. Combining these DC vaccines with Osimertinib offers a promising and safe treatment for EGFRm NSCLC.
Collapse
Affiliation(s)
- Yao Li
- Medical School of Chinese People’s Liberation Army (PLA), Beijing 100000, China; (Y.L.); (X.G.); (P.Z.); (T.L.); (H.F.)
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100000, China;
| | - Yaping Long
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100000, China;
- Institute of Oncology, The First Medical Center of Chinese, PLA General Hospital, Beijing 100000, China;
| | - Xiangwei Ge
- Medical School of Chinese People’s Liberation Army (PLA), Beijing 100000, China; (Y.L.); (X.G.); (P.Z.); (T.L.); (H.F.)
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100000, China;
| | - Pengfei Zhang
- Medical School of Chinese People’s Liberation Army (PLA), Beijing 100000, China; (Y.L.); (X.G.); (P.Z.); (T.L.); (H.F.)
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100000, China;
| | - Tao Li
- Medical School of Chinese People’s Liberation Army (PLA), Beijing 100000, China; (Y.L.); (X.G.); (P.Z.); (T.L.); (H.F.)
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100000, China;
| | - Liangliang Wu
- Institute of Oncology, The First Medical Center of Chinese, PLA General Hospital, Beijing 100000, China;
| | - Hao Fan
- Medical School of Chinese People’s Liberation Army (PLA), Beijing 100000, China; (Y.L.); (X.G.); (P.Z.); (T.L.); (H.F.)
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100000, China;
| | - Zhijuan Du
- Medical School of Chinese People’s Liberation Army (PLA), Beijing 100000, China; (Y.L.); (X.G.); (P.Z.); (T.L.); (H.F.)
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100000, China;
| | - Qiaowei Liu
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100000, China;
- Institute of Oncology, The First Medical Center of Chinese, PLA General Hospital, Beijing 100000, China;
| | - Yi Hu
- Medical School of Chinese People’s Liberation Army (PLA), Beijing 100000, China; (Y.L.); (X.G.); (P.Z.); (T.L.); (H.F.)
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100000, China;
- Institute of Oncology, The First Medical Center of Chinese, PLA General Hospital, Beijing 100000, China;
| |
Collapse
|
9
|
Vučemilović A. Exosomes: intriguing mediators of intercellular communication in the organism's response to noxious agents. Arh Hig Rada Toksikol 2024; 75:228-239. [PMID: 39718095 PMCID: PMC11667715 DOI: 10.2478/aiht-2024-75-3923] [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: 11/01/2024] [Revised: 11/01/2024] [Accepted: 12/01/2024] [Indexed: 12/25/2024] Open
Abstract
Exosomes are small extracellular vesicles that range from 30 to 150 nm in size and are formed through cellular endocytosis. They consist of proteins, lipids, and nucleic acids at varying ratios and quantities. The composition and spatiotemporal dynamics of exosomes suggest that they play a crucial role in intercellular communication. The information conveyed by exosomes significantly impacts the regulation of health and disease states in the organism. The term "noxious" refers to all harmful environmental agents and conditions that can disrupt the physiological equilibrium and induce pathological states, regardless whether of radiological, biological, or chemical origin. This review comprehensively examines the presence of such noxious agents within the organism in relation to exosome formation and function. Furthermore, it explores the cause-effect relationship between noxious agents and exosomes, aiming to restore physiological homeostasis and prepare the organism for defence against harmful agents. Regardless of the specific bioinformatic content associated with each noxious agent, synthesis of data on the interactions between various types of noxious agents and exosomes reveals that an organized defence against these agents is unachievable without the support of exosomes. Consequently, exosomes are identified as the primary communication and information system within an organism, with their content being pivotal in maintaining the health-disease balance.
Collapse
|
10
|
Liang C, Wang M, Huang Y, Yam JWP, Zhang X, Zhang X. Recent Advances of Small Extracellular Vesicles for the Regulation and Function of Cancer-Associated Fibroblasts. Int J Mol Sci 2024; 25:12548. [PMID: 39684264 DOI: 10.3390/ijms252312548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous cell population in the tumor microenvironment (TME) that critically affect cancer progression. Small extracellular vesicles (sEVs) act as information messengers by transmitting a wide spectrum of biological molecules, including proteins, nucleic acids, and metabolites, from donor cells to recipient cells. Previous studies have demonstrated that CAFs play important roles in tumor progression by regulating tumor cell proliferation, metastasis, therapeutic resistance, and metabolism via sEVs. In turn, tumor-derived sEVs can also regulate the activation and phenotype switch of CAFs. The dynamic crosstalk between CAFs and cancer cells via sEVs could ultimately determine cancer progression. In this review, we summarized the recent advance of the biological roles and underlying mechanisms of sEVs in mediating CAF-tumor cell interaction and its impact on cancer progression. We also reviewed the clinical applications of tumor- and CAF-derived sEVs, which could identify novel potential targets and biomarkers for cancer diagnosis, therapy, and prognosis.
Collapse
Affiliation(s)
- Chengdong Liang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Maoye Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yongli Huang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Xiaoxin Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
11
|
Youssef A, Sahgal A, Das S. Radioresistance and brain metastases: a review of the literature and applied perspective. Front Oncol 2024; 14:1477448. [PMID: 39540151 PMCID: PMC11557554 DOI: 10.3389/fonc.2024.1477448] [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: 08/08/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Intracranial metastatic disease is a serious complication of cancer, treated through surgery, radiation, and targeted therapies. The central role of radiation therapy makes understanding the radioresistance of metastases a priori a key interest for prognostication and therapeutic development. Although historically defined clinic-radiographically according to tumour response, developments in new techniques for delivering radiation treatment and understanding of radioprotective mechanisms led to a need to revisit the definition of radioresistance in the modern era. Factors influencing radioresistance include tumour-related factors (hypoxia, cancer stem cells, tumour kinetics, tumour microenvironment, metabolic alterations, tumour heterogeneity DNA damage repair, non-coding RNA, exosomes, methylomes, and autophagy), host-related factors (volume effect & dose-limiting non-cancerous tissue, pathophysiology, and exosomes), technical factors, and probabilistic factors (cell cycle and random gravity of DNA damage). Influences on radioresistance are introduced and discussed in the context of brain metastases.
Collapse
Affiliation(s)
- Andrew Youssef
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Hospital, Toronto, ON, Canada
| | - Sunit Das
- Division of Neurosurgery, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
12
|
Lai Z, Ye T, Zhang M, Mu Y. Exosomes as Vehicles for Noncoding RNA in Modulating Inflammation: A Promising Regulatory Approach for Ischemic Stroke and Myocardial Infarction. J Inflamm Res 2024; 17:7485-7501. [PMID: 39464334 PMCID: PMC11505480 DOI: 10.2147/jir.s484119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
Exosomes have grown as promising carriers for noncoding RNAs (ncRNAs) in the treatment of inflammation, particularly in conditions like ischemic stroke and myocardial infarction. These ncRNAs, which include microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), play a crucial role in regulating inflammatory pathways, presenting new therapeutic opportunities. In both ischemic stroke and myocardial infarction, inflammation significantly influences disease progression and severity. Exosomes can deliver ncRNAs directly to specific cells and tissues, providing a targeted approach to modulate gene expression and reduce inflammation. Their biocompatibility and low risk of inducing immune responses make exosomes ideal therapeutic vehicles. Ongoing research is focused on optimizing the loading of ncRNAs into exosomes, ensuring efficient delivery, and understanding the mechanisms by which these ncRNAs mitigate inflammation. In ischemic stroke, exosome-derived ncRNAs originate from various cell types, including neurons, M2 microglia, patient serum, genetically engineered HEK293T cells, and mesenchymal stromal cells. In the case of myocardial infarction, these ncRNAs are sourced from mesenchymal stem cells, endothelial cells, and patient plasma. These exosome-loaded ncRNAs play a significant role in modulating inflammation in both ischemic stroke and myocardial infarction. As this research advances, therapies based on exosomes may completely change how diseases linked to inflammation are treated, offering new avenues for patient care and recovery. This review explores the latest advancements in understanding how exosomes impact specific inflammatory components, with a particular emphasis on the role of ncRNAs contained in exosomes. The review concludes by highlighting the clinical potential of exosome-derived ncRNAs as innovative therapeutic and diagnostic tools.
Collapse
Affiliation(s)
- Zhuhong Lai
- Department of Cardiology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, People’s Republic of China
| | - Tingqiao Ye
- Department of Cardiology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, People’s Republic of China
| | - Mingjun Zhang
- Department of Cardiology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, People’s Republic of China
| | - Ying Mu
- Department of Cardiology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, People’s Republic of China
| |
Collapse
|
13
|
Ahmad R, Barcellini A, Baumann K, Benje M, Bender T, Bragado P, Charalampopoulou A, Chowdhury R, Davis AJ, Ebner DK, Eley J, Kloeber JA, Mutter RW, Friedrich T, Gutierrez-Uzquiza A, Helm A, Ibáñez-Moragues M, Iturri L, Jansen J, Morcillo MÁ, Puerta D, Kokko AP, Sánchez-Parcerisa D, Scifoni E, Shimokawa T, Sokol O, Story MD, Thariat J, Tinganelli W, Tommasino F, Vandevoorde C, von Neubeck C. Particle Beam Radiobiology Status and Challenges: A PTCOG Radiobiology Subcommittee Report. Int J Part Ther 2024; 13:100626. [PMID: 39258166 PMCID: PMC11386331 DOI: 10.1016/j.ijpt.2024.100626] [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: 07/03/2024] [Accepted: 08/02/2024] [Indexed: 09/12/2024] Open
Abstract
Particle therapy (PT) represents a significant advancement in cancer treatment, precisely targeting tumor cells while sparing surrounding healthy tissues thanks to the unique depth-dose profiles of the charged particles. Furthermore, their linear energy transfer and relative biological effectiveness enhance their capability to treat radioresistant tumors, including hypoxic ones. Over the years, extensive research has paved the way for PT's clinical application, and current efforts aim to refine its efficacy and precision, minimizing the toxicities. In this regard, radiobiology research is evolving toward integrating biotechnology to advance drug discovery and radiation therapy optimization. This shift from basic radiobiology to understanding the molecular mechanisms of PT aims to expand the therapeutic window through innovative dose delivery regimens and combined therapy approaches. This review, written by over 30 contributors from various countries, provides a comprehensive look at key research areas and new developments in PT radiobiology, emphasizing the innovations and techniques transforming the field, ranging from the radiobiology of new irradiation modalities to multimodal radiation therapy and modeling efforts. We highlight both advancements and knowledge gaps, with the aim of improving the understanding and application of PT in oncology.
Collapse
Affiliation(s)
- Reem Ahmad
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Amelia Barcellini
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- Clinical Department Radiation Oncology Unit, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Kilian Baumann
- Institute of Medical Physics and Radiation Protection, University of Applied Sciences Giessen, Giessen, Germany
- Marburg Ion-Beam Therapy Center, Marburg, Germany
| | - Malte Benje
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Tamara Bender
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Paloma Bragado
- Biochemistry and Molecular Biology Department, Complutense University of Madrid, Madrid, Spain
| | - Alexandra Charalampopoulou
- University School for Advanced Studies (IUSS), Pavia, Italy
- Radiobiology Unit, Development and Research Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Reema Chowdhury
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Anthony J. Davis
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel K. Ebner
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - John Eley
- Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jake A. Kloeber
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert W. Mutter
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas Friedrich
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | | | - Alexander Helm
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Marta Ibáñez-Moragues
- Medical Applications of Ionizing Radiation Unit, Technology Department, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Lorea Iturri
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Jeannette Jansen
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Miguel Ángel Morcillo
- Medical Applications of Ionizing Radiation Unit, Technology Department, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Daniel Puerta
- Departamento de Física Atómica, Molecular y Nuclear, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Complejo Hospitalario Universitario de Granada/Universidad de Granada, Granada, Spain
| | | | | | - Emanuele Scifoni
- TIFPA-INFN - Trento Institute for Fundamental Physics and Applications, Trento, Italy
| | - Takashi Shimokawa
- National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Olga Sokol
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | | | - Juliette Thariat
- Centre François Baclesse, Université de Caen Normandie, ENSICAEN, CNRS/IN2P3, LPC Caen UMR6534, Caen, France
| | - Walter Tinganelli
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Francesco Tommasino
- TIFPA-INFN - Trento Institute for Fundamental Physics and Applications, Trento, Italy
- Department of Physics, University of Trento, Trento, Italy
| | - Charlot Vandevoorde
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Cläre von Neubeck
- Department of Particle Therapy, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| |
Collapse
|
14
|
Jassi C, kuo WW, Kuo CH, Chang CM, Chen MC, Shih TC, Li CC, Huang CY. Mediation of radiation-induced bystander effect and epigenetic modification: The role of exosomes in cancer radioresistance. Heliyon 2024; 10:e34460. [PMID: 39114003 PMCID: PMC11304029 DOI: 10.1016/j.heliyon.2024.e34460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/20/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Exosomes are nano-sized extracellular vesicles produced by almost all mammalian cells. They play an important role in cell-to-cell communication by transferring biologically active molecules from the cell of origin to the recipient cells. Ionizing radiation influences exosome production and molecular cargo loading. In cancer management, ionizing radiation is a form of treatment that exerts its cancer cytotoxicity by induction of DNA damage and other alterations to the targeted tissue cells. However, normal bystander non-targeted cells may exhibit the effects of ionizing radiation, a phenomenon called radiation-induced bystander effect (RIBE). The mutual communication between the two groups of cells (targeted and non-targeted) via radiation-influenced exosomes enables the exchange of radiosensitive molecules. This facilitates indirect radiation exposure, leading, among other effects, to epigenetic remodeling and subsequent adaptation to radiation. This review discusses the role exosomes play in epigenetically induced radiotherapy resistance through the mediation of RIBE.
Collapse
Affiliation(s)
- Chikondi Jassi
- Department of Biological Sciences and Technology, China Medical University, Taichung, Taiwan
| | - Wei-Wen kuo
- Department of Biological Sciences and Technology, China Medical University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
- Department of Kinesiology and Health Science, College of William and Mary, Williamsburg, VA, USA
| | - Chun-Ming Chang
- Department of General Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine Tzu Chi University, 701, Section 3, Chung-Yang Road, Hualien 97004, Taiwan
| | - Ming-Cheng Chen
- Division of Colorectal Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tzu-Ching Shih
- Department of Biomedical Imaging & Radiological Science College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Chi-Cheng Li
- School of Medicine Tzu Chi University, 701, Section 3, Chung-Yang Road, Hualien 97004, Taiwan
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Center of Stem Cell & Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondria Related Diseases Research Center, Hualien Tzu Chi Hospital, Hualien 970, Taiwan
- Graduate Institute of Biomedicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung 413, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
15
|
Wu Y, Cao Y, Chen L, Lai X, Zhang S, Wang S. Role of Exosomes in Cancer and Aptamer-Modified Exosomes as a Promising Platform for Cancer Targeted Therapy. Biol Proced Online 2024; 26:15. [PMID: 38802766 PMCID: PMC11129508 DOI: 10.1186/s12575-024-00245-2] [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: 04/14/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
Exosomes are increasingly recognized as important mediators of intercellular communication in cancer biology. Exosomes can be derived from cancer cells as well as cellular components in tumor microenvironment. After secretion, the exosomes carrying a wide range of bioactive cargos can be ingested by local or distant recipient cells. The released cargos act through a variety of mechanisms to elicit multiple biological effects and impact most if not all hallmarks of cancer. Moreover, owing to their excellent biocompatibility and capability of being easily engineered or modified, exosomes are currently exploited as a promising platform for cancer targeted therapy. In this review, we first summarize the current knowledge of roles of exosomes in risk and etiology, initiation and progression of cancer, as well as their underlying molecular mechanisms. The aptamer-modified exosome as a promising platform for cancer targeted therapy is then briefly introduced. We also discuss the future directions for emerging roles of exosome in tumor biology and perspective of aptamer-modified exosomes in cancer therapy.
Collapse
Affiliation(s)
- Yating Wu
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China
- Department of Medical Oncology, Fuzhou General Clinical Medical School (the 900th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Yue Cao
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Li Chen
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Xiaofeng Lai
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Shenghang Zhang
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China.
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China.
| | - Shuiliang Wang
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China.
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China.
| |
Collapse
|
16
|
Zafra J, Onieva JL, Oliver J, Garrido-Barros M, González-Hernández A, Martínez-Gálvez B, Román A, Ordóñez-Marmolejo R, Pérez-Ruiz E, Benítez JC, Mesas A, Vera A, Chicas-Sett R, Rueda-Domínguez A, Barragán I. Novel Blood Biomarkers for Response Prediction and Monitoring of Stereotactic Ablative Radiotherapy and Immunotherapy in Metastatic Oligoprogressive Lung Cancer. Int J Mol Sci 2024; 25:4533. [PMID: 38674117 PMCID: PMC11050102 DOI: 10.3390/ijms25084533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Up to 80% of patients under immune checkpoint inhibitors (ICI) face resistance. In this context, stereotactic ablative radiotherapy (SABR) can induce an immune or abscopal response. However, its molecular determinants remain unknown. We present early results of a translational study assessing biomarkers of response to combined ICI and SABR (I-SABR) in liquid biopsy from oligoprogressive patients in a prospective observational multicenter study. Cohort A includes metastatic patients in oligoprogression to ICI maintaining the same ICI due to clinical benefit and who receive concomitant SABR. B is a comparative group of oligometastatic patients receiving only SABR. Blood samples are extracted at baseline (T1), after the first (T2) and last (T3) fraction, two months post-SABR (T4) and at further progression (TP). Response is evaluated by iRECIST and defined by the objective response rate (ORR)-complete and partial responses. We assess peripheral blood mononuclear cells (PBMCs), circulating cell-free DNA (cfDNA) and small RNA from extracellular vesicles. Twenty-seven patients could be analyzed (cohort A: n = 19; B: n = 8). Most were males with non-small cell lung cancer and one progressing lesion. With a median follow-up of 6 months, the last ORR was 63% (26% complete and 37% partial response). A decrease in cfDNA from T2 to T3 correlated with a good response. At T2, CD8+PD1+ and CD8+PDL1+ cells were increased in non-responders and responders, respectively. At T2, 27 microRNAs were differentially expressed. These are potential biomarkers of response to I-SABR in oligoprogressive disease.
Collapse
Affiliation(s)
- Juan Zafra
- Group of Translational Research in Cancer Immunotherapy (CIMO2), Department of Radiation Oncology, Virgen de la Victoria University Hospital, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain;
- Faculty of Medicine, University of Malaga (UMA), 29071 Málaga, Spain; (J.L.O.); (M.G.-B.); (A.G.-H.)
| | - Juan Luis Onieva
- Faculty of Medicine, University of Malaga (UMA), 29071 Málaga, Spain; (J.L.O.); (M.G.-B.); (A.G.-H.)
- Group of Translational Research in Cancer Immunotherapy (CIMO2), Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria Hospitals, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain; (J.O.); (B.M.-G.); (E.P.-R.); (J.C.B.)
| | - Javier Oliver
- Group of Translational Research in Cancer Immunotherapy (CIMO2), Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria Hospitals, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain; (J.O.); (B.M.-G.); (E.P.-R.); (J.C.B.)
| | - María Garrido-Barros
- Faculty of Medicine, University of Malaga (UMA), 29071 Málaga, Spain; (J.L.O.); (M.G.-B.); (A.G.-H.)
- Group of Translational Research in Cancer Immunotherapy (CIMO2), Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria Hospitals, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain; (J.O.); (B.M.-G.); (E.P.-R.); (J.C.B.)
| | - Andrea González-Hernández
- Faculty of Medicine, University of Malaga (UMA), 29071 Málaga, Spain; (J.L.O.); (M.G.-B.); (A.G.-H.)
- Group of Translational Research in Cancer Immunotherapy (CIMO2), Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria Hospitals, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain; (J.O.); (B.M.-G.); (E.P.-R.); (J.C.B.)
| | - Beatriz Martínez-Gálvez
- Group of Translational Research in Cancer Immunotherapy (CIMO2), Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria Hospitals, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain; (J.O.); (B.M.-G.); (E.P.-R.); (J.C.B.)
| | - Alicia Román
- Department of Radiation Oncology, Virgen de la Victoria University Hospital, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain; (A.R.); (R.O.-M.)
| | - Rafael Ordóñez-Marmolejo
- Department of Radiation Oncology, Virgen de la Victoria University Hospital, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain; (A.R.); (R.O.-M.)
| | - Elisabeth Pérez-Ruiz
- Group of Translational Research in Cancer Immunotherapy (CIMO2), Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria Hospitals, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain; (J.O.); (B.M.-G.); (E.P.-R.); (J.C.B.)
| | - José Carlos Benítez
- Group of Translational Research in Cancer Immunotherapy (CIMO2), Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria Hospitals, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain; (J.O.); (B.M.-G.); (E.P.-R.); (J.C.B.)
| | - Andrés Mesas
- Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria Hospitals, 29010 Málaga, Spain;
| | - Andrés Vera
- Department of Radiation Oncology, Dr Negrín University Hospital, 35010 Las Palmas de Gran Canaria, Spain;
| | - Rodolfo Chicas-Sett
- Department of Radiation Oncology, La Fe University Hospital, 46026 Valencia, Spain;
- Group of Clinical and Translational Cancer Research, Le Fe Health Research Institute, 46026 Valencia, Spain
| | - Antonio Rueda-Domínguez
- Group of Translational Research in Cancer Immunotherapy (CIMO2), Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria Hospitals, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain; (J.O.); (B.M.-G.); (E.P.-R.); (J.C.B.)
| | - Isabel Barragán
- Group of Translational Research in Cancer Immunotherapy (CIMO2), Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria Hospitals, Institute of Biomedical Research in Malaga (IBIMA), 29010 Málaga, Spain; (J.O.); (B.M.-G.); (E.P.-R.); (J.C.B.)
- Group of Pharmacoepigenetics, Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
17
|
Robinson SD, Samuels M, Jones W, Gilbert D, Critchley G, Giamas G. Shooting the messenger: a systematic review investigating extracellular vesicle isolation and characterisation methods and their influence on understanding extracellular vesicles-radiotherapy interactions in glioblastoma. BMC Cancer 2023; 23:939. [PMID: 37798728 PMCID: PMC10552223 DOI: 10.1186/s12885-023-11437-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) hold promise for improving our understanding of radiotherapy response in glioblastoma due to their role in intercellular communication within the tumour microenvironment (TME). However, methodologies to study EVs are evolving with significant variation within the EV research community. METHODS We conducted a systematic review to critically appraise EV isolation and characterisation methodologies and how this influences our understanding of the findings from studies investigating radiotherapy and EV interactions in glioblastoma. 246 articles published up to 24/07/2023 from PubMed and Web of Science were identified using search parameters related to radiotherapy, EVs, and glioblastoma. Two reviewers evaluated study eligibility and abstracted data. RESULTS In 26 articles eligible for inclusion (16 investigating the effects of radiotherapy on EVs, five investigating the effect of EVs on radiation response, and five clinical studies), significant heterogeneity and frequent omission of key characterisation steps was identified, reducing confidence that the results are related to EVs and their cargo as opposed to co-isolated bioactive molecules. However, the results are able to clearly identify interactions between EVs and radiotherapy bi-directionally within different cell types within the glioblastoma TME. These interactions facilitate transferable radioresistance and oncogenic signalling, highlighting that EVs are an important component in the variability of glioblastoma radiotherapy response. CONCLUSIONS Future multi-directional investigations interrogating the whole TME are required to improve subsequent clinical translation, and all studies should incorporate up to date controls and reporting requirements to increase the validity of their findings. This would be facilitated by increased collaboration between less experienced and more experienced EV research groups.
Collapse
Affiliation(s)
- Stephen David Robinson
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, John Maynard Smith Building, Falmer, Brighton, BN1 9QG, UK, (SDR, MS, WJ, GG).
- Sussex Cancer Centre, University Hospitals Sussex NHS Foundation Trust, Brighton, UK, (SDR, DG).
| | - Mark Samuels
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, John Maynard Smith Building, Falmer, Brighton, BN1 9QG, UK, (SDR, MS, WJ, GG)
| | - William Jones
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, John Maynard Smith Building, Falmer, Brighton, BN1 9QG, UK, (SDR, MS, WJ, GG)
| | - Duncan Gilbert
- Sussex Cancer Centre, University Hospitals Sussex NHS Foundation Trust, Brighton, UK, (SDR, DG)
- Medical Research Council Clinical Trials Unit, University College London, London, UK, (DG)
| | - Giles Critchley
- Department of Neurosurgery, University Hospitals Sussex NHS Foundation Trust, Brighton, UK, (GC)
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, John Maynard Smith Building, Falmer, Brighton, BN1 9QG, UK, (SDR, MS, WJ, GG)
| |
Collapse
|
18
|
Zhao D, Mo Y, Neganova ME, Aleksandrova Y, Tse E, Chubarev VN, Fan R, Sukocheva OA, Liu J. Dual effects of radiotherapy on tumor microenvironment and its contribution towards the development of resistance to immunotherapy in gastrointestinal and thoracic cancers. Front Cell Dev Biol 2023; 11:1266537. [PMID: 37849740 PMCID: PMC10577389 DOI: 10.3389/fcell.2023.1266537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023] Open
Abstract
Successful clinical methods for tumor elimination include a combination of surgical resection, radiotherapy, and chemotherapy. Radiotherapy is one of the crucial components of the cancer treatment regimens which allow to extend patient life expectancy. Current cutting-edge radiotherapy research is focused on the identification of methods that should increase cancer cell sensitivity to radiation and activate anti-cancer immunity mechanisms. Radiation treatment activates various cells of the tumor microenvironment (TME) and impacts tumor growth, angiogenesis, and anti-cancer immunity. Radiotherapy was shown to regulate signaling and anti-cancer functions of various TME immune and vasculature cell components, including tumor-associated macrophages, dendritic cells, endothelial cells, cancer-associated fibroblasts (CAFs), natural killers, and other T cell subsets. Dual effects of radiation, including metastasis-promoting effects and activation of oxidative stress, have been detected, suggesting that radiotherapy triggers heterogeneous targets. In this review, we critically discuss the activation of TME and angiogenesis during radiotherapy which is used to strengthen the effects of novel immunotherapy. Intracellular, genetic, and epigenetic mechanisms of signaling and clinical manipulations of immune responses and oxidative stress by radiotherapy are accented. Current findings indicate that radiotherapy should be considered as a supporting instrument for immunotherapy to limit the cancer-promoting effects of TME. To increase cancer-free survival rates, it is recommended to combine personalized radiation therapy methods with TME-targeting drugs, including immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Deyao Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingyi Mo
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Margarita E. Neganova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russia
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Chernogolovka, Russia
| | - Yulia Aleksandrova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russia
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Chernogolovka, Russia
| | - Edmund Tse
- Department of Hepatology, Royal Adelaide Hospital, CALHN, Adelaide, SA, Australia
| | - Vladimir N. Chubarev
- Sechenov First Moscow State Medical University, Sechenov University, Moscow, Russia
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Olga A. Sukocheva
- Department of Hepatology, Royal Adelaide Hospital, CALHN, Adelaide, SA, Australia
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Pasqualetti F, Miniati M, Gonnelli A, Gadducci G, Giannini N, Palagini L, Mancino M, Fuentes T, Paiar F. Cancer Stem Cells and Glioblastoma: Time for Innovative Biomarkers of Radio-Resistance? BIOLOGY 2023; 12:1295. [PMID: 37887005 PMCID: PMC10604498 DOI: 10.3390/biology12101295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/28/2023]
Abstract
Despite countless papers in the field of radioresistance, researchers are still far from clearly understanding the mechanisms triggered in glioblastoma. Cancer stem cells (CSC) are important to the growth and spread of cancer, according to many studies. In addition, more recently, it has been suggested that CSCs have an impact on glioblastoma patients' prognosis, tumor aggressiveness, and treatment outcomes. In reviewing this new area of biology, we will provide a summary of the most recent research on CSCs and their role in the response to radio-chemotherapy in GB. In this review, we will examine the radiosensitivity of stem cells. Moreover, we summarize the current knowledge of the biomarkers of stemness and evaluate their potential function in the study of radiosensitivity.
Collapse
Affiliation(s)
- Francesco Pasqualetti
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56100 Pisa, Italy; (F.P.); (A.G.); (G.G.); (N.G.); (M.M.); (T.F.); (F.P.)
| | - Mario Miniati
- Department of Clinical and Experimental Medicine, University of Pisa, Italy, Via Roma 67, 56100 Pisa, Italy;
| | - Alessandra Gonnelli
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56100 Pisa, Italy; (F.P.); (A.G.); (G.G.); (N.G.); (M.M.); (T.F.); (F.P.)
| | - Giovanni Gadducci
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56100 Pisa, Italy; (F.P.); (A.G.); (G.G.); (N.G.); (M.M.); (T.F.); (F.P.)
| | - Noemi Giannini
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56100 Pisa, Italy; (F.P.); (A.G.); (G.G.); (N.G.); (M.M.); (T.F.); (F.P.)
| | - Laura Palagini
- Department of Clinical and Experimental Medicine, University of Pisa, Italy, Via Roma 67, 56100 Pisa, Italy;
| | - Maricia Mancino
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56100 Pisa, Italy; (F.P.); (A.G.); (G.G.); (N.G.); (M.M.); (T.F.); (F.P.)
| | - Taiusha Fuentes
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56100 Pisa, Italy; (F.P.); (A.G.); (G.G.); (N.G.); (M.M.); (T.F.); (F.P.)
| | - Fabiola Paiar
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56100 Pisa, Italy; (F.P.); (A.G.); (G.G.); (N.G.); (M.M.); (T.F.); (F.P.)
| |
Collapse
|
20
|
Tang JY, Chuang YT, Shiau JP, Yen CY, Chang FR, Tsai YH, Farooqi AA, Chang HW. Connection between Radiation-Regulating Functions of Natural Products and miRNAs Targeting Radiomodulation and Exosome Biogenesis. Int J Mol Sci 2023; 24:12449. [PMID: 37569824 PMCID: PMC10419287 DOI: 10.3390/ijms241512449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/29/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Exosomes are cell-derived membranous structures primarily involved in the delivery of the payload to the recipient cells, and they play central roles in carcinogenesis and metastasis. Radiotherapy is a common cancer treatment that occasionally generates exosomal miRNA-associated modulation to regulate the therapeutic anticancer function and side effects. Combining radiotherapy and natural products may modulate the radioprotective and radiosensitizing responses of non-cancer and cancer cells, but there is a knowledge gap regarding the connection of this combined treatment with exosomal miRNAs and their downstream targets for radiation and exosome biogenesis. This review focuses on radioprotective natural products in terms of their impacts on exosomal miRNAs to target radiation-modulating and exosome biogenesis (secretion and assembly) genes. Several natural products have individually demonstrated radioprotective and miRNA-modulating effects. However, the impact of natural-product-modulated miRNAs on radiation response and exosome biogenesis remains unclear. In this review, by searching through PubMed/Google Scholar, available reports on potential functions that show radioprotection for non-cancer tissues and radiosensitization for cancer among these natural-product-modulated miRNAs were assessed. Next, by accessing the miRNA database (miRDB), the predicted targets of the radiation- and exosome biogenesis-modulating genes from the Gene Ontology database (MGI) were retrieved bioinformatically based on these miRNAs. Moreover, the target-centric analysis showed that several natural products share the same miRNAs and targets to regulate radiation response and exosome biogenesis. As a result, the miRNA-radiomodulation (radioprotection and radiosensitization)-exosome biogenesis axis in regard to natural-product-mediated radiotherapeutic effects is well organized. This review focuses on natural products and their regulating effects on miRNAs to assess the potential impacts of radiomodulation and exosome biogenesis for both the radiosensitization of cancer cells and the radioprotection of non-cancer cells.
Collapse
Affiliation(s)
- Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ya-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (F.-R.C.); (Y.-H.T.)
| | - Yi-Hong Tsai
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (F.-R.C.); (Y.-H.T.)
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan
| | - Hsueh-Wei Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| |
Collapse
|
21
|
Averbeck D. Low-Dose Non-Targeted Effects and Mitochondrial Control. Int J Mol Sci 2023; 24:11460. [PMID: 37511215 PMCID: PMC10380638 DOI: 10.3390/ijms241411460] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Non-targeted effects (NTE) have been generally regarded as a low-dose ionizing radiation (IR) phenomenon. Recently, regarding long distant abscopal effects have also been observed at high doses of IR) relevant to antitumor radiation therapy. IR is inducing NTE involving intracellular and extracellular signaling, which may lead to short-ranging bystander effects and distant long-ranging extracellular signaling abscopal effects. Internal and "spontaneous" cellular stress is mostly due to metabolic oxidative stress involving mitochondrial energy production (ATP) through oxidative phosphorylation and/or anaerobic pathways accompanied by the leakage of O2- and other radicals from mitochondria during normal or increased cellular energy requirements or to mitochondrial dysfunction. Among external stressors, ionizing radiation (IR) has been shown to very rapidly perturb mitochondrial functions, leading to increased energy supply demands and to ROS/NOS production. Depending on the dose, this affects all types of cell constituents, including DNA, RNA, amino acids, proteins, and membranes, perturbing normal inner cell organization and function, and forcing cells to reorganize the intracellular metabolism and the network of organelles. The reorganization implies intracellular cytoplasmic-nuclear shuttling of important proteins, activation of autophagy, and mitophagy, as well as induction of cell cycle arrest, DNA repair, apoptosis, and senescence. It also includes reprogramming of mitochondrial metabolism as well as genetic and epigenetic control of the expression of genes and proteins in order to ensure cell and tissue survival. At low doses of IR, directly irradiated cells may already exert non-targeted effects (NTE) involving the release of molecular mediators, such as radicals, cytokines, DNA fragments, small RNAs, and proteins (sometimes in the form of extracellular vehicles or exosomes), which can induce damage of unirradiated neighboring bystander or distant (abscopal) cells as well as immune responses. Such non-targeted effects (NTE) are contributing to low-dose phenomena, such as hormesis, adaptive responses, low-dose hypersensitivity, and genomic instability, and they are also promoting suppression and/or activation of immune cells. All of these are parts of the main defense systems of cells and tissues, including IR-induced innate and adaptive immune responses. The present review is focused on the prominent role of mitochondria in these processes, which are determinants of cell survival and anti-tumor RT.
Collapse
Affiliation(s)
- Dietrich Averbeck
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France
| |
Collapse
|
22
|
Peng J, Yin X, Yun W, Meng X, Huang Z. Radiotherapy-induced tumor physical microenvironment remodeling to overcome immunotherapy resistance. Cancer Lett 2023; 559:216108. [PMID: 36863506 DOI: 10.1016/j.canlet.2023.216108] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/14/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023]
Abstract
The clinical benefits of immunotherapy are proven in many cancers, but a significant number of patients do not respond well to immunotherapy. The tumor physical microenvironment (TpME) has recently been shown to affect the growth, metastasis and treatment of solid tumors. The tumor microenvironment (TME) has unique physical hallmarks: 1) unique tissue microarchitecture, 2) increased stiffness, 3) elevated solid stress, and 4) elevated interstitial fluid pressure (IFP), which contribute to tumor progression and immunotherapy resistance in a variety of ways. Radiotherapy, a traditional and powerful treatment, can remodel the matrix and blood flow associated with the tumor to improve the response rate of immune checkpoint inhibitors (ICIs) to a certain extent. Herein, we first review the recent research advances on the physical properties of the TME and then explain how TpME is involved in immunotherapy resistance. Finally, we discuss how radiotherapy can remodel TpME to overcome immunotherapy resistance.
Collapse
Affiliation(s)
- Jianfeng Peng
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Wenhua Yun
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| | - Zhaoqin Huang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|