1
|
Zeng JQ, Gao YW, Jia XB. Harnessing artificial intelligence to address immune response heterogeneity in low-dose radiation therapy. World J Radiol 2025; 17:108011. [DOI: 10.4329/wjr.v17.i5.108011] [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: 04/02/2025] [Revised: 04/12/2025] [Accepted: 05/08/2025] [Indexed: 05/26/2025] Open
Abstract
Low-dose radiation therapy has emerged as a promising modality for cancer treatment because of its ability to stimulate antitumor immune responses while minimizing damage to healthy tissues. However, the significant heterogeneity in immune responses among patients complicates its clinical application, hindering outcome prediction and treatment personalization. Artificial intelligence (AI) offers a transformative solution by integrating multidimensional data such as immunomics, radiomics, and clinical features to decode complex immune patterns and predict individual therapeutic outcomes. This editorial explored the potential of AI to address immune response heterogeneity in low-dose radiation therapy and proposed an AI-driven framework for precision immunotherapy. While promising, challenges, including data standardization, model interpretability, and clinical validation, must be overcome to ensure successful integration into oncological practice.
Collapse
Affiliation(s)
- Jing-Qi Zeng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, China
| | - Yi-Wei Gao
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, China
| | - Xiao-Bin Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, China
| |
Collapse
|
2
|
Dong J, Qi Y, Sha S, Fu C, Xu X, Li B. Whole-body CT scanning radiation improves the immune microenvironment of tumor tissues to enhance the antitumor effect of ICI. BMC Cancer 2025; 25:824. [PMID: 40316957 PMCID: PMC12049032 DOI: 10.1186/s12885-025-14119-7] [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: 05/16/2024] [Accepted: 04/08/2025] [Indexed: 05/04/2025] Open
Abstract
OBJECTIVE The effect of frequent whole-body CT scans during immune checkpoint inhibitor (ICI) therapy on patients' anti-tumor immunity. METHODS We conducted a retrospective clinical study aimed to investigate the correlation between the frequency of CT scans during immune checkpoint inhibitor (ICI) therapy and the duration of remission (DOR) of ICI therapy in patients with stage IV non-small cell lung cancer (NSCLC). We constructed a hormonal mouse model and administered immune checkpoint inhibitor (ICI) therapy to mice, and radiated five whole-body CT scans to mice during ICI therapy to observe whether frequent whole-body CT scans had an effect on the antitumor effect of immunotherapy in mice. RESULTS The more frequent CT scans during patients' immune checkpoint inhibitor (ICI) treatment the longer the duration of remission (DOR) of ICI treatment. In a mouse model we observed that the addition of whole-body CT scanning radiation had a tendency to inhibit tumor growth in mice compared with the anti-PD-1 group alone.Frequent CT scanning radiation during the application of immune checkpoint inhibitor PD-1 increased the proportion of infiltrating CD8 + T cells in tumor tissues and significantly increased the proportion of IFNγ-secreting CD8 + T cells, and single-cell sequencing of the results also revealed that IFNγ and killing-related genes were significantly upregulated in tumor-infiltrating CD8T cells. CONCLUSION To our knowledge this is the first study on the effect of CT scan radiation on ICI.Our findings suggest that multiple CT scans during immune checkpoint inhibitor (ICI) treatment did not promote tumor progression, but instead a trend toward delayed tumor progression was observed.
Collapse
Affiliation(s)
- Jigang Dong
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300000, China
- Qingdao People's Hospital Group (Jiaozhou), Jiaozhou Central Hospital of Qingdao, Qingdao, China
| | - Ying Qi
- Qingdao People's Hospital Group (Jiaozhou), Jiaozhou Central Hospital of Qingdao, Qingdao, China
| | - Sha Sha
- Qingdao People's Hospital Group (Jiaozhou), Jiaozhou Central Hospital of Qingdao, Qingdao, China
| | - Chengrui Fu
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300000, China
- Department of Radiotherapy, Shandong Cancer Hospital, Jinan, 250000, China
| | - Xiao Xu
- Qingdao People's Hospital Group (Jiaozhou), Jiaozhou Central Hospital of Qingdao, Qingdao, China.
| | - Baosheng Li
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300000, China.
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Department of Radiation Oncology, Tianjin, China.
- Medical University; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong, Jinan, China.
- Academy of Medical Sciences, No. 440 Jiyan Road, Jinan, 250017, China.
| |
Collapse
|
3
|
Cao W, Su K, Lu C, Li J, Gui X. Effect and mechanism of the miR-1284/EIF4A1 axis on the cGAS-STING pathway under radiotherapy. Transl Cancer Res 2025; 14:2483-2494. [PMID: 40386253 PMCID: PMC12079598 DOI: 10.21037/tcr-2025-603] [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: 03/17/2025] [Accepted: 04/10/2025] [Indexed: 05/20/2025]
Abstract
Background Gastric cancer (GC) remains a major global health concern, with limited treatment options, especially in advanced stages. Radiotherapy (RT) plays a vital role in GC management, but resistance to DNA damage impedes its effectiveness. MicroRNA-1284 (miR-1284), a tumor suppressor, regulates eukaryotic translation initiation factor 4A1 (EIF4A1), which is involved in DNA damage repair through homologous recombination (HR). This axis has been implicated in enhancing GC cell survival following RT. Additionally, the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, activated by DNA damage, plays a key role in triggering an anti-tumor immune response. However, the interaction between the miR-1284/EIF4A1 axis, DNA repair, and the cGAS-STING pathway in GC under RT conditions remains unclear. This study aims to investigate how the miR-1284/EIF4A1 axis influences DNA repair and its role in activating the cGAS-STING pathway to enhance RT efficacy in GC. Methods A stably expressed messenger miR-1284 cell line was established. Quantitative reverse transcription and western blot were used to examine the expression of miR-1284 and EIF4A1, and the effect of blocking the miR-1284/EIF4A1 axis on the cGAS-STING pathway and interferon-β (IFN-β) in GC cells after RT; cytotoxicity experiments were conducted to explore the mechanism of the miR-1284/EIF4A1 axis in radiation-induced DNA damage repair; animal experiments were conducted to explore the translational application of rocaglamide (RocA) combined with the programmed cell death-ligand 1 (PD-L1) antibody in RT. Results The miR-1284/EIF4A1 axis in the GC cells promoted the repair of radiation-induced DNA damage and was associated with the prognosis of GC patients. Blocking this axis delayed the C-terminal binding protein interacting protein (CtIP)-mediated DNA repair, enhanced RT effectiveness, and activated the cGAS-STING pathway, while increasing the rate of apoptosis. In vivo experiments based on RocA binding to PD-L1 antibodies under RT had good biological safety, and thus provide a potential therapeutic strategy for the treatment of GC. Conclusions The miR-1284/EIF4A1 axis promotes the repair of DNA damage caused by RT, promotes the activation of the cGAS-STING pathway in GC, and has good biological safety. Our findings provide an important experimental basis for enhancing the anti-tumor immune effect of RT in the treatment of GC.
Collapse
Affiliation(s)
- Wenlong Cao
- Department of Gastrointestine and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ka Su
- Department of Gastrointestine and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chunmiao Lu
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiehua Li
- Department of Gastrointestine and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaolong Gui
- Department of Gastrointestine and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
4
|
Dong J, Fu C, Li M, Wang Z, Li B. Radiation from frequent whole-body CT scans induces systemic immunosuppression and immune activation of tumor tissue. Transl Oncol 2025; 54:102326. [PMID: 40014978 PMCID: PMC11909445 DOI: 10.1016/j.tranon.2025.102326] [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: 08/19/2024] [Revised: 12/30/2024] [Accepted: 02/13/2025] [Indexed: 03/01/2025] Open
Abstract
OBJECTIVE This study aims to elucidate the impact of repeated whole-body computed tomography (CT) scans on systemic immunity, the tumor immune microenvironment, and tumor control. This inquiry was prompted by clinical observations indicating a decrease in the levels of IFN-β and IFN-γ in patients' blood following whole-body CT scans. METHODS A Lewis lung carcinoma (LLC) mouse model was established and divided into two groups: a control group and a group subjected to multiple whole-body CT scanning radiation (WBCTSs). The study monitored tumor growth trends across both groups and employed a comprehensive set of analytical techniques-including enzyme-linked immunosorbent assay (ELISA), flow cytometry analysis, immunohistochemistry, RNA sequencing, and single-cell sequencing-to assess differences in cytokine profiles (IFN-β and IFN-γ), proportions of key immune cells, and gene expression variations between the groups. RESULTS Repeated CT scan radiation does not promote tumor progression. In tumor tissues subjected to multiple CT scans, an increase in the proportion of CD8+ T cells, elevated interferon levels, and up-regulation of genes associated with killing in CD8+ T cells and genes associated with Ifnb in macrophages were observed. In contrast, radiation from multiple whole-body CT scans resulted in a decrease in the proportion of CD8+ T cells in the blood and spleen, a decrease in serum interferon levels, and down-regulation of killing-related genes in CD8+ T cells. CONCLUSION Our results suggest that repeated whole-body CT scanning radiation induces systemic immunosuppression and immune activation in tumor tissues. Multiple repeated CT scans do not promote tumor progression.
Collapse
Affiliation(s)
- Jigang Dong
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300000, China; Qingdao People's Hospital Group (Jiaozhou), Jiaozhou Central Hospital of Qingdao. China.
| | - Chengrui Fu
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300000, China; Department of Radiotherapy. Shandong Cancer Hospital, Jinan, 250000. China
| | - Minghao Li
- Department of Radiotherapy. Shandong Cancer Hospital, Jinan, 250000. China
| | - Zhongtang Wang
- Department of Radiotherapy. Shandong Cancer Hospital, Jinan, 250000. China
| | - Baosheng Li
- Department of Radiotherapy, Shandong Cancer Hospital, Jinan, China.
| |
Collapse
|
5
|
Rafiq Z, Kang M, Barsoumian HB, Manzar GS, Hu Y, Leuschner C, Huang A, Masrorpour F, Lu W, Puebla-Osorio N, Welsh JW. Enhancing immunotherapy efficacy with synergistic low-dose radiation in metastatic melanoma: current insights and prospects. J Exp Clin Cancer Res 2025; 44:31. [PMID: 39881333 PMCID: PMC11781074 DOI: 10.1186/s13046-025-03281-2] [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: 09/25/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
Recent advances in oncology research have highlighted the promising synergy between low-dose radiation therapy (LDRT) and immunotherapies, with growing evidence highlighting the unique benefits of the combination. LDRT has emerged as a potent tool for stimulating the immune system, triggering systemic antitumor effects by remodeling the tumor microenvironment. Notably, LDRT demonstrates remarkable efficacy even in challenging metastatic sites such as the liver (uveal) and brain (cutaneous), particularly in advanced melanoma stages. The increasing interest in utilizing LDRT for secondary metastatic sites of uveal, mucosal, or cutaneous melanomas underscores its potential efficacy in combination with various immunotherapies. This comprehensive review traverses the journey from laboratory research to clinical applications, elucidating LDRT's immunomodulatory role on the tumor immune microenvironment (TIME) and systemic immune responses. We meticulously examine the preclinical evidence and ongoing clinical trials, throwing light on the promising prospects of LDRT as a complementary therapy in melanoma treatment. Furthermore, we explore the challenges associated with LDRT's integration into combination therapies, addressing crucial factors such as optimal dosage, fractionation, treatment frequency, and synergy with other pharmacological agents. Considering its low toxicity profile, LDRT presents a compelling case for application across multiple lesions, augmenting the antitumor immune response in poly-metastatic disease scenarios. The convergence of LDRT with other disciplines holds immense potential for developing novel radiotherapy-combined modalities, paving the way for more effective and personalized treatment strategies in melanoma and beyond. Moreover, the dose-related toxicities of immunotherapies may be reduced by synergistic amplification of antitumor efficacy with LDRT.
Collapse
Affiliation(s)
- Zahid Rafiq
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79968, USA
| | - Mingyo Kang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gohar S Manzar
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yun Hu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Carola Leuschner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ailing Huang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Fatemeh Masrorpour
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Weiqin Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79968, USA
| | - Nahum Puebla-Osorio
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
6
|
Chen CQ, Huang H, Pan M, Jia Z, Zhang J, Chen QQ. Combination therapy of low-dose radiotherapy and immunotherapy in advanced metastatic nasopharyngeal carcinoma: a case report and literature review. Discov Oncol 2025; 16:52. [PMID: 39812951 PMCID: PMC11735707 DOI: 10.1007/s12672-025-01794-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Nasopharyngeal cancer (NPC) is a common head and neck malignant tumor, which is difficult to treat at the advanced NPC due to its occult and high metastatic potential to the cervical lymph nodes and distant organs. Low-dose radiotherapy (LDRT) is increasingly being investigated for potential cancer treatment. When combined with immune checkpoint inhibitors, LDRT has been shown to significantly improve the immune microenvironment of tumors, thereby promote the immune attack on tumor cells. However, the therapeutic effect of LDRT combined with immunotherapy in advanced NPC is not well understood. We report a case of a 31-year-old man was diagnosed with advanced metastatic nasopharnygeal non-keratinizing carcinoma (T4N3M1 stage IVb AJCC8th). The patient was treated with a high-dose of radiation therapy combined with LDRT and immunotherapy to inhibit tumor cell proliferation and activate the body's immune system. The initial treatment procedure was as follows: chemotherapy regimen (nedaplatin + docetaxel + fluorouracil) induction, followed by radical radiotherapy for the primary lesion, LDRT for the L5 vertebral body metastasis, and concurrent use of low-dose capecitabine beat chemotherapy and toripalimab immunotherapy. The patient was also administered with human granulocyte-macrophage colony-stimulating factor and aspirin to enhance the immune function. This combination therapy approach alleviated patient symptoms, improved bone changes in the L5 vertebral body and resolved the tumor without any adverse effects signals. The progression-free survival (PFS) has reached 27 months and he is currently stable without tumor recurrence. CONCLUSION The combination of chemotherapy and LDRT with aspirin and human granulocyte macrophage colony-stimulating factor improved the disease state of advanced NPC cancer, effectively reducing the level of tumor markers, enhanced the immune function without significant adverse reactions.
Collapse
Affiliation(s)
- Chun-Qiao Chen
- Department of Oncology, People's Hospital of Guilin, No. 12 Wenming Road, Guilin, 541002, Guangxi Zhuang Autonomous Region, China
| | - Hui Huang
- Department of Oncology, People's Hospital of Guilin, No. 12 Wenming Road, Guilin, 541002, Guangxi Zhuang Autonomous Region, China
| | - Min Pan
- Department of Oncology, People's Hospital of Guilin, No. 12 Wenming Road, Guilin, 541002, Guangxi Zhuang Autonomous Region, China
| | - Zhe Jia
- Department of Oncology, People's Hospital of Guilin, No. 12 Wenming Road, Guilin, 541002, Guangxi Zhuang Autonomous Region, China
| | - Jing Zhang
- Department of Oncology, People's Hospital of Guilin, No. 12 Wenming Road, Guilin, 541002, Guangxi Zhuang Autonomous Region, China
| | - Qiu-Qiu Chen
- Department of Oncology, People's Hospital of Guilin, No. 12 Wenming Road, Guilin, 541002, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
7
|
Cao Z, Deng K, Jiang J, Tian K, Wang B. Combined treatment of small cell lung cancer using radiotherapy and immunotherapy: Challenges and updates. Biomed Pharmacother 2025; 182:117727. [PMID: 39675137 DOI: 10.1016/j.biopha.2024.117727] [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: 09/18/2024] [Revised: 11/17/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024] Open
Abstract
Currently, chemotherapy remains the standard first- and second-line treatment for small cell lung cancer (SCLC). Research concerning immunotherapy has brought about a remarkable development in the treatment pattern of SCLC. Atirizumab, duvalizumab, atezolizumab, and serplulimab can significantly improve the clinical outcomes of SCLC. Given the rapidly evolving concept that combining immunotherapy with radiotherapy can increase therapeutic effectiveness, clinicians are devoted to further improving local tumor control by integrating immunotherapy with radiotherapy. This paper reviews the research progress in this field to date and explores ways to further enhance the efficacy of this combination therapy. We first discussed that immunotherapy combined with radiotherapy can improve the abscopal effect, progression-free survival, and overall survival rates of SCLC patients. Then, the biomarkers related to the radiation immune microenvironment, such as programmed death ligand-1 (PD-L1), tumor mutational burden (TMB), and the immune function of patients were discussed. Next, we explored the occurrence and underlying mechanisms of immune resistance during radiotherapy implementation. Finally, we clarified that the emerging trend of low-dose radiotherapy help overcome the inhibitory signals that limit T-cell infiltration in the tumor matrix. In summary, considering the rapid development of this field, these combined therapy strategies may have unlimited potential to further improve the efficacy of radiotherapy combined with immunotherapy for patients.
Collapse
Affiliation(s)
- Zhumin Cao
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing 400054, China.
| | - Kai Deng
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing 400061, China.
| | - Jinxiu Jiang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing 400061, China.
| | - Ke Tian
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing 400054, China.
| | - Bin Wang
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing 400054, China.
| |
Collapse
|
8
|
Scott BR. Radiophobia Harm, Its Main Cause, and a Proposed Solution. Dose Response 2025; 23:15593258251318305. [PMID: 40160708 PMCID: PMC11951894 DOI: 10.1177/15593258251318305] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/18/2024] [Accepted: 01/13/2025] [Indexed: 04/02/2025] Open
Abstract
Background: We are exposed to natural ionizing radiation and other genomic stressors throughout life and radiophobia has caused much harm to society. The main basis for radiophobia is the invalid linear no-threshold (LNT) hypothesis for cancer induction, which the System of Radiological Protection (SRP) is linked to. Largely unknown to the public, evolution-associated genomic stress adaptation (gensadaptation) over many previous generations now provides protection to all lifeforms from low radiation doses. Objective: To help bring about an improved SRP not linked to the invalid LNT hypothesis for radiation-caused health detriment and to promote low-dose radiation therapy for different diseases. Methods: All-solid-cancer mortality risk dose-response relationships for A-bomb survivors were generated based on published LNT-modeling-related results. Dose-response relationships for lung cancer prevention by low-dose radiation were generated by linear interpolation based on published data from a study using > 15,000 mice. Uncertainty characterization was based on Monte Carlo calculations for binomial and Poisson distributions. New dose characterization tools were used for threshold dose-response relationships for radiation-caused cancer mortality. Results: The all-solid-cancer mortality risk for A-bomb survivors transitioned from LNT to threshold-linear when adjusted for key missing uncertainty at low doses. The prevention of lung cancer in mice by low radiation doses depends on the radiation absorbed dose and type. Conclusions: The SRP should be linked to population dose thresholds rather than the invalid LNT hypothesis and small likely harmless radiation doses could possibly be used in treating different diseases.
Collapse
Affiliation(s)
- Bobby R. Scott
- Lovelace Biomedical Research Institute, Albuquerque, NM, USA
| |
Collapse
|
9
|
Yao X, Huo W, Wang Y, Xia D, Chen Y, Tang Y, Tang H, Yang W, Liu Y, Xue J, Yuan Q, Gao X, Cao K. Environmental Low-Dose Radiation Activates Th1 Immunity through the Mitochondria-STING Pathway. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:22907-22918. [PMID: 39689952 DOI: 10.1021/acs.est.4c08009] [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: 12/19/2024]
Abstract
The presence of low-dose radiation (LDR) in the environment has become more prevalent. However, the effect of LDR exposure on the immune system remains elusive. Here, we interestingly found that LDR specifically elevated the percentage of CD4+IFNγ+ Th1 splenocytes, both in vitro and in vivo, without affecting the percentage of CD8+IFNγ+ Tc1 cells and regulatory T cells. A similar phenomenon was found in T cells from peripheral blood. Mechanistically, we found that LDR can induce mitochondrial damage, which stimulated the STING signaling pathway, leading to the enhanced expression of T-bet, the master transcriptional factor of Th1-cell differentiation. The specific STING signal inhibitor can abrogate the effect of LDR on Th1 differentiation, confirming the central role of the STING pathway. To further validate the immunoregulatory role of LDR, we exposed mice with whole body LDR and evaluated if LDR could protect mice against triple-negative breast cancer through enhanced antitumor immunity. As expected, LDR significantly delayed tumor development and promoted cell death. Meanwhile, LDR resulted in increased tumor-infiltrating Th1 cells, while the proportion of Tc1 and Treg cells remained unchanged. Furthermore, the infiltration of antitumor macrophages was also increased. In summary, we revealed that environmental LDR could specifically regulate Th1 T-cell activities, providing critical information for the potential application of LDR in both clinical and nonclinical settings.
Collapse
Affiliation(s)
- Xiuxiu Yao
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Wendi Huo
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Yuchen Wang
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Dongfang Xia
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Yan Chen
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Yuhua Tang
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Huayong Tang
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Wenjiang Yang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Liu
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Jingquan Xue
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Yuan
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Xueyun Gao
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Kai Cao
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
10
|
Shao N, Yang Y, Hu G, Luo Q, Cheng N, Chen J, Huang Y, Zhang H, Luo L, Xiao Z. Synergistic enhancement of low-dose radiation therapy via cuproptosis and metabolic reprogramming for radiosensitization in in situ hepatocellular carcinoma. J Nanobiotechnology 2024; 22:772. [PMID: 39696547 DOI: 10.1186/s12951-024-03011-4] [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: 09/23/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Radiotherapy (RT) is a primary clinical approach for cancer treatment, but its efficacy is often hindered by various challenges, especially radiation resistance, which greatly compromises the therapeutic effectiveness of RT. Mitochondria, central to cellular energy metabolism and regulation of cell death, play a critical role in mechanisms of radioresistance. In this context, cuproptosis, a novel copper-induced mitochondria-respiratory-dependent cell death pathway, offers a promising avenue for radiosensitization. RESULTS In this study, an innovative theranostic nanoplatform was designed to induce cuproptosis in synergy with low-dose radiation therapy (LDRT, i.e., 0.5-2 Gy) for the treatment of in situ hepatocellular carcinoma (HCC). This approach aims to reverse the hypoxic tumor microenvironment, promoting a shift in cellular metabolism from glycolysis to oxidative phosphorylation (OXPHOS), thereby enhancing sensitivity to cuproptosis. Concurrently, the Fenton-like reaction ensures a sustained supply of copper and depletion of glutathione (GSH), inducing cuproptosis, disrupting mitochondrial function, and interrupting the energy supply. This strategy effectively overcomes radioresistance and enhances the therapeutic efficacy against tumors. CONCLUSIONS In conclusion, this study elucidates the intricate interactions among tumor hypoxia reversal, cuproptosis, metabolic reprogramming, and radiosensitization, particularly in the context of treating in situ hepatocellular carcinoma, thereby providing a novel paradigm for radiotherapy.
Collapse
Affiliation(s)
- Ni Shao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yongqing Yang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Genwen Hu
- Department of Radiology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Qiao Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Nianlan Cheng
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jifeng Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Hong Zhang
- Department of Interventional Radiology and Vascular Surgery, The Sixth Affiliated Hospital of Jinan University, Dongguan, 523067, China.
| | - Liangping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
- Department of Radiology and Nuclear Medicine, The Fifth Affiliated Hospital of Jinan University (Shenhe People's Hospital), Heyuan, 517000, China.
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
11
|
Ganina A, Askarov M, Kozina L, Karimova M, Shayakhmetov Y, Mukhamedzhanova P, Brimova A, Berikbol D, Chuvakova E, Zaripova L, Baigenzhin A. Prospects for Treatment of Lung Cancer Using Activated Lymphocytes Combined with Other Anti-Cancer Modalities. Adv Respir Med 2024; 92:504-525. [PMID: 39727496 PMCID: PMC11673795 DOI: 10.3390/arm92060045] [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: 10/18/2024] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
This review explores the significance and prospects of using diverse T-cell variants in the context of combined therapy for lung cancer treatment. Recently, there has been an increase in research focused on understanding the critical role of tumor-specific T lymphocytes and the potential benefits of autologous T-cell-based treatments for individuals with lung cancer. One promising approach involves intravenous administration of ex vivo-activated autologous lymphocytes to improve the immune status of patients with cancer. Investigations are also exploring the factors that influence the success of T-cell therapy and the methods used to stimulate them. Achieving a comprehensive understanding of the characteristics of activated lymphocytes and deciphering the mechanisms underlying their activation of innate anti-tumor immunity will pave the way for numerous clinical trials and the development of innovative strategies for cancer therapy like combined immunotherapy and radiation therapy.
Collapse
Affiliation(s)
- Anastasia Ganina
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Manarbek Askarov
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Larissa Kozina
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Madina Karimova
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Yerzhan Shayakhmetov
- International Oncological Tomotherapy Center “YMIT”, Astana 010009, Kazakhstan; (Y.S.); (P.M.); (A.B.); (D.B.)
| | - Perizat Mukhamedzhanova
- International Oncological Tomotherapy Center “YMIT”, Astana 010009, Kazakhstan; (Y.S.); (P.M.); (A.B.); (D.B.)
| | - Aigul Brimova
- International Oncological Tomotherapy Center “YMIT”, Astana 010009, Kazakhstan; (Y.S.); (P.M.); (A.B.); (D.B.)
| | - Daulet Berikbol
- International Oncological Tomotherapy Center “YMIT”, Astana 010009, Kazakhstan; (Y.S.); (P.M.); (A.B.); (D.B.)
| | - Elmira Chuvakova
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Lina Zaripova
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Abay Baigenzhin
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| |
Collapse
|
12
|
Looi CK, Loo EM, Lim HC, Chew YL, Chin KY, Cheah SC, Goh BH, Mai CW. Revolutionizing the treatment for nasopharyngeal cancer: the impact, challenges and strategies of stem cell and genetically engineered cell therapies. Front Immunol 2024; 15:1484535. [PMID: 39450176 PMCID: PMC11499120 DOI: 10.3389/fimmu.2024.1484535] [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: 08/22/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a distinct malignancy of the nasopharynx and is consistently associated with the Epstein-Barr virus (EBV) infection. Its unique anatomical location and complex aetiology often result in advanced-stage disease at first diagnosis. While radiotherapy (RT) and chemotherapy have been the mainstays of treatment, they often fail to prevent tumour recurrence and metastasis, leading to high rates of treatment failure and mortality. Recent advancement in cell-based therapies, such as chimeric antigen receptor (CAR)-T cell therapy, have shown great promise in hematological malignancies and are now being investigated for NPC. However, challenges such as targeting specific tumour antigens, limited T cell persistence and proliferation, and managing treatment-related toxicities must be addressed. Extensive research is needed to enhance the effectiveness and safety of these therapies, paving the way for their integration into standard clinical practice for better management of NPC and a better quality of life for human health.
Collapse
Affiliation(s)
- Chin-King Looi
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Ee-Mun Loo
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
- Advanced Genomics Laboratory, AGTC Genomics, Kuala Lumpur, Malaysia
| | - Heng-Chee Lim
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Yik-Ling Chew
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shiau-Chuen Cheah
- Faculty of Medicine and Health Sciences, UCSI University, Port Dickson, Negeri Sembilan, Malaysia
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre, School of Medical and Life Sciences, Sunway University Malaysia, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, China
| | - Chun-Wai Mai
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Wang D, Huang L, Qian D, Cao Y, Wu X, Xu P, Ming L, Tang J, Huang Z, Yin Y, Zhou L. Low-dose radiotherapy promotes the formation of tertiary lymphoid structures in lung adenocarcinoma. Front Immunol 2024; 14:1334408. [PMID: 38259481 PMCID: PMC10800908 DOI: 10.3389/fimmu.2023.1334408] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
PURPOSE A tertiary lymphoid structure (TLS) refers to an organized infiltration of immune cells that is linked to a positive prognosis and improved response to immunotherapy. However, methods that promote TLS formation are limited and challenging to implement in clinical settings. In this study, we aimed to promote the formation and maturation of TLSs in lung adenocarcinoma (LUAD) by combining low-dose radiotherapy (LDRT) with immunotherapy. METHODS Tissue sections from 198 patients who had undergone surgery were examined. Risk factors for patient survival were assessed, and the relationship between TLSs and five-year survival was analyzed. The Kras-LSL-G12D spontaneous lung cancer mouse model was used to screen the optimal irradiation dose (0/1/2 Gy whole lung irradiation) for promoting TLS formation. LDRT combined with anti-PD-1 was used to promote the formation and maturation of TLSs. RESULTS TLS+, TLSHigh, TLS+GC+ and CD8High within TLS+ were associated with a favorable prognosis. LDRT increased the formation of early TLSs in the Kras-LSL-G12D lung cancer mouse model. In addition, LDRT combined with anti-PD-1 treatment can significantly improve the maturity of TLSs in mouse LUAD, resulting in greater antitumor effects. This antitumor effect was strongly associated with the number of CD8+ T cells within the TLSs. CONCLUSION We successfully applied LDRT combined with PD-1 inhibitor therapy for the first time, which increased both the quantity and maturity of TLSs in lung cancer. This approach achieved a promising antitumor effect.
Collapse
Affiliation(s)
- Duo Wang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Liuying Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Danqi Qian
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yulin Cao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaohan Wu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Peiwen Xu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Liang Ming
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Junhui Tang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yuan Yin
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Leyuan Zhou
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
- Department of Radiation Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
14
|
Frosina G. Radiotherapy of high-grade gliomas: dealing with a stalemate. Crit Rev Oncol Hematol 2023; 190:104110. [PMID: 37657520 DOI: 10.1016/j.critrevonc.2023.104110] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/14/2023] [Accepted: 08/23/2023] [Indexed: 09/03/2023] Open
Abstract
This article discusses the studies on radiotherapy of high-grade gliomas published between January 1, 2022, and June 30, 2022, with special reference to their molecular biology basis. The focus was on advances in radioresistance, radiosensitization and the toxicity of radiotherapy treatments. In the first half of 2022, several important advances have been made in understanding resistance mechanisms in high-grade gliomas. Furthermore, the development of several radiosensitization procedures for these deadly tumors, including studies with small molecule radiosensitizers, new fractionation protocols, and new immunostimulatory agents, has progressed in both the preclinical and clinical settings, reflecting the frantic research effort in the field. However, since 2005 our research efforts fail to produce significant improvements to treatment guidelines for high-grade gliomas. Possible reasons for this stalemate and measures to overcome it are discussed.
Collapse
Affiliation(s)
- Guido Frosina
- Mutagenesis & Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy.
| |
Collapse
|