1
|
Yin Z, Zhang H, Zhang K, Yue J, Tang R, Wang Y, Deng Q, Yu Q. Impacts of combining PD-L1 inhibitor and radiotherapy on the tumour immune microenvironment in a mouse model of esophageal squamous cell carcinoma. BMC Cancer 2025; 25:474. [PMID: 40087599 PMCID: PMC11909915 DOI: 10.1186/s12885-025-13801-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND The combination of radiation with immune checkpoint inhibitors (ICIs) has been demonstrated to display synergistic effects in solid cancers. Nevertheless, the anti-tumor effect of combining radiation with programmed cell death 1 ligand 1 (PD-L1) inhibitor in esophageal squamous cell carcinoma (ESCC) has remained unclear. Therefore, the objectives of our study were to evaluate the anti-tumor effects of PD-L1 inhibitors combined with radiotherapy in a mouse model of ESCC and to depict the immune landscape within the tumor microenvironment (TME). METHODS Murine ESCC cells (mEC25) were injected subcutaneously into the right flanks of C57BL/6 mice. Tumor-bearing mice were exposed to different treatments: IgG antibody (control), anti-PD-L1 antibody, radiation, or radiation + anti-PD-L1 antibody. Tumor growth and survival time of mice were monitored. Tumour immune microenvironment was assessed by flow cytometry, including CD4+T cells, CD8+T cells, regulatory T cells (Tregs), tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and the activation and exhaustion of CD8+T cell. In addition, transcriptomic analysis was used to examine the changes in immune gene expression in the TME. RESULTS Radiotherapy combined with anti-PD-L1 inhibitors (radioimmunotherapy) synergistically enhanced anti-tumor immune response, leading to decreased tumor growth and prolonged survival of tumor-bearing mice. The radioimmunotherapy increased the infiltration of CD8+ T cells, the ratio of CD8+ T cells to Tregs, the population of central memory CD8+ T cells (TCM), interferon-gamma (IFN-γ) secretion of tumor-infiltrating CD8+ T cells, and reduced the accumulation of M2-type TAMs and Tregs in the TME in mouse model. In addition, the radioimmunotherapy induced anti-tumor immune response in the spleen and tumor-draining lymph node (TDLN). Moreover, transcriptomic analysis suggested that the radioimmunotherapy promoted the activation of immune regulatory pathways and increased the expression of cytokines such as CXCL9 and CXCL10, thus creating an immunoinflammatory tumor microenvironment. CONCLUSIONS Our research revealed that anti-PD-L1 inhibitors combined with radiotherapy caused systemic anti-tumor immunity by reshaping the immune microenvironment in a mouse model of ESCC.
Collapse
Affiliation(s)
- Zihao Yin
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Hongfang Zhang
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Ke Zhang
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Jing Yue
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Rongjun Tang
- Hyperthermia Center, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Yaping Wang
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
| | - Qinghua Deng
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China.
| | - Qingqing Yu
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Hangzhou, 310002, China.
| |
Collapse
|
2
|
Zhang YP, Guo ZQ, Cai XT, Rong ZX, Fang Y, Chen JQ, Zhuang KM, Ruan MJ, Ma SC, Lin LY, Han DD, Li YS, Wang YY, Wang J, Cao CH, Tang XR, Xie QK, Chen Y, Lin Y, Tan JL, Yu ZH, Wu ZN, Wei W, Zheng DY, Zeng YJ, Ruan YC, Xu ZP, Gu JZ, Xiao LS, Liu L, Guan J, Bai X, Wu DH, Dong ZY. PAI-1-driven SFRP2 high cancer-associated fibroblasts hijack the abscopal effect of radioimmunotherapy. Cancer Cell 2025:S1535-6108(25)00076-5. [PMID: 40086438 DOI: 10.1016/j.ccell.2025.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/15/2024] [Accepted: 02/20/2025] [Indexed: 03/16/2025]
Abstract
The abscopal effect of radioimmunotherapy, wherein tumor shrinkage occurs beyond the irradiated field, is therapeutically promising but clinically rare. The mechanisms underlying this effect remain elusive. Here, in vivo genome-wide CRISPR screening identifies SFRP2 as a potential stromal regulator of the abscopal effect. SFRP2 exhibits cancer-associated fibroblast (CAF)-specific expression and radioimmunotherapy-mediated upregulation in unirradiated tumors. Conditional Sfrp2 knockout in CAFs boosts the abscopal effect by rewiring the vascular-immune microenvironment to promote CD8+ T cell recruitment to unirradiated tumors. In vivo lineage tracing reveals that elevated SFRP2 correlates with radioimmunotherapy-driven pericyte lineage commitment. Serum proteomics reveals that irradiated-tumor-secreted PAI-1 triggers distant tumor pericyte cell-fate transition into SFRP2high CAFs via the LRP1/p65 axis. Pharmacologically blocking SFRP2 or PAI-1 enhances the abscopal effect in humanized patient-derived xenograft models. Our findings collectively illustrate that PAI-1-induced SFRP2high CAFs serve as critical stromal regulator to hijack the abscopal effect, providing promising targets for enhancing radioimmunotherapy effectiveness.
Collapse
Affiliation(s)
- Yan-Pei Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ze-Qin Guo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Ting Cai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zi-Xuan Rong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuan Fang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jia-Qi Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kui-Mao Zhuang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Min-Jie Ruan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Si-Cong Ma
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Le-Yi Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Duan-Duan Han
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yang-Si Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yuan-Yuan Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jian Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chuan-Hui Cao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xin-Ran Tang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qian-Kun Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yue Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yan Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jia-Le Tan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zi-Hang Yu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ze-Nan Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wei Wei
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, Hubei 441000, China
| | - Da-Yong Zheng
- Department of Oncology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Shunde 528333, China
| | - Yu-Jie Zeng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Ying-Chen Ruan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zi-Peng Xu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jun-Zi Gu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lu-Shan Xiao
- Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Li Liu
- Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Health Management Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jian Guan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Xue Bai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - De-Hua Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Zhong-Yi Dong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
3
|
Xu C, Zhou GQ, Li WF, Hu DS, Chen XZ, Lin SJ, Jin F, Huang XQ, Peng G, Huang J, Wu Y, Tao CJ, Li JB, Lin AH, Zhao HY, Hong SB, Huang HL, Tang LL, Peng YL, Shi KF, Chen L, Qi LP, Yang KY, Shen LF, Sun Y, Ma J. Nivolumab combined with induction chemotherapy and radiotherapy in nasopharyngeal carcinoma: A multicenter phase 2 PLATINUM trial. Cancer Cell 2025:S1535-6108(25)00032-7. [PMID: 40020668 DOI: 10.1016/j.ccell.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/13/2024] [Accepted: 01/31/2025] [Indexed: 03/03/2025]
Abstract
Severe toxicities caused by concurrent cisplatin are a critical problem in nasopharyngeal carcinoma (NPC) treatment. In this phase 2 multicenter PLATINUM trial (NCT03984357), we recruited 152 NPC patients who received 12-cycle nivolumab plus induction chemotherapy and radiotherapy without concurrent cisplatin. After a median follow-up of 43 months, the 3-year failure-free survival (FFS) was 88.5% (95% confidence interval [CI], 83.4%-93.8%) and the 3-year overall survival was 97.9%. An early clearance of Epstein-Barr virus (EBV) DNA after induction-phase treatment was associated with FFS benefit. Sixty (40.2%) and eight (5.2%) patients had acute and late grade 3-4 adverse events (AEs), respectively. Most patients had good tolerance to AE-associated frequency (68.0%-96.7%), severity (56.0%-98.6%), and interference (58.0%-98.0%); 86.7%-100.0% of quality-of-life domains showed either no clinically meaningful deterioration or a rapid recovery. Nivolumab plus induction chemotherapy and radiotherapy demonstrated efficacious anti-tumor activity, low toxicity, and favorable tolerability and quality-of-life for NPC patients.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China
| | - Guan-Qun Zhou
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China
| | - Wen-Fei Li
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China
| | - De-Sheng Hu
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Xiao-Zhong Chen
- Department of Head and Neck Tumor Radiotherapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, P.R. China
| | - Shao-Jun Lin
- Department of Radiation Oncology, Cancer Hospital of Fujian Medical University (Fujian Provincial Cancer Hospital), Fuzhou, Fujian 350014, P.R. China
| | - Feng Jin
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Xin-Qiong Huang
- Department of Radiation Oncology, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Gang Peng
- Department of Oncology, Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jing Huang
- Department of Oncology, Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yuan Wu
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Chang-Juan Tao
- Department of Head and Neck Tumor Radiotherapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, P.R. China
| | - Ji-Bin Li
- Clinical Trials Center, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510000, P.R. China
| | - Ai-Hua Lin
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Hong-Yun Zhao
- Department of Medical Oncology, and Department of Clinical Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510000, P.R. China
| | - Shu-Bin Hong
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Hui-Ling Huang
- Department of Cardiology, Cardiac Prevention and Assessment Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Ling-Long Tang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China
| | - Ying-Lin Peng
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China
| | - Ke-Fu Shi
- Nursing Division, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510000, P.R. China
| | - Liu Chen
- Nursing Division, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510000, P.R. China
| | - Li-Ping Qi
- Nursing Division, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510000, P.R. China
| | - Kun-Yu Yang
- Department of Oncology, Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China.
| | - Liang-Fang Shen
- Department of Radiation Oncology, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China.
| | - Ying Sun
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China; Chinese Society of Clinical Oncology, Beijing 100000, P.R. China.
| | - Jun Ma
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China; Chinese Society of Clinical Oncology, Beijing 100000, P.R. China.
| |
Collapse
|
4
|
Huang L, Wang D, Xu M, Qian D, Cao Y, Wu X, Ming L, Tang J, Huang Z, Yin Y, Zhou L. Mixed radiation with different doses induces CCL17 to recruit CD8 +T cell to exert anti-tumor effects in non-small cell lung cancer. Front Immunol 2025; 15:1508007. [PMID: 39877375 PMCID: PMC11772420 DOI: 10.3389/fimmu.2024.1508007] [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: 10/08/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Background Different doses of radiotherapy (RT) exert diverse effects on tumor immunity, although the precise irradiation method remains unknown. This study sought to elucidate the influence of combining different doses of RT with immune checkpoint inhibitors (ICIs) on the infiltration of CD8+T cells within tumors, thereby augmenting the anti-tumor response. Methods Constructing a mouse model featuring bilateral lung cancer tumors subjected to high and low dose irradiation, the analysis of RNA transcriptome sequencing data and immunohistochemical validation for tumors exposed to various dosages guided the selection of the optimal low-dose irradiation scheme. Subsequently, upon the integration of immune checkpoint inhibitors (ICIs) therapy, the infiltration of immune cells within the tumor was ascertained via immunohistochemistry (IHC) and flow cytometry (FCM). Finally, through bioinformatics analysis and experimental verification, potential strategies to bolster the anti-tumor immune response were investigated. Results In comparison to the administration of 20Gy alone to the primary tumor, supplementing with 6Gy directed at the abscopal tumor produces a more pronounced abscopal response. The synergy of 20Gy, 6Gy, and ICIs markedly boosts the efficiency of ICIs. According to the findings from IHC and FCM studies, the triple therapy group exhibits a heightened infiltration of immune cells into the tumor, largely attributable to the augmented expression of CCL17 within the tumor under these irradiation regimens, which subsequently draws CD8+ T cells to infiltrate the tumor site, exerting cytotoxic effects. Conclusion Our study shows that the combined application of 20Gy and 6Gy can enhance the infiltration of tumor CD8+T cells in mice and improve the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Liuying Huang
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Duo Wang
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Muchen Xu
- Department of Radiation Oncology, The Fourth Affiliated Hospital of Soochow University, Suzhou, 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
| | - 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, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
5
|
Surgo A, Davì V, Ciliberti MP, Carbonara R, Caliandro M, Di Guglielmo FC, Sasso N, Calbi R, Gentile MA, Talienti T, Bruno I, Troia M, Bonaparte I, Ludovico GM, Surico G, Fiorentino A. Metabolic Complete Response of Metastatic Oncogene-Negative, PDL1-Negative Non-Small Cell Lung Cancer After Chemo-Immunotherapy and Radiotherapy: A Case Report. Curr Oncol 2024; 31:8118-8126. [PMID: 39727721 DOI: 10.3390/curroncol31120598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024] Open
Abstract
A 71-year-old male ex-smoker presented in October 2021 to our department with a brain and bone metastatic adenocarcinoma NSCLC. PDL1, ROS, EGFR, and ALK were negative. He underwent stereotactic radiotherapy for brain metastases. In November 2021, he started a chemotherapy (CHT) regimen with cisplatin (75 mg/m2 every 21 days) and pemetrexed (500 mg/m2 every 21 days), and ICI with Atezolizumab (1200 mg every 21 days). In July 2022, RT to the lung tumor and mediastinal nodal was performed with a total dose of 45 Gy in 15 fractions. He continued with immunotherapy until December 2022, when a grade 3-4 toxicity from immunotherapy was observed (hypothyroidism, psoriasis, and cystitis). He achieved a complete clinical response to the therapy. To date, the patient is alive, with a complete metabolic response, without treatment at 37 months from diagnosis.
Collapse
Affiliation(s)
- Alessia Surgo
- Radiation Oncology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, BA, Italy
| | - Valerio Davì
- Radiation Oncology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Maria Paola Ciliberti
- Radiation Oncology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Roberta Carbonara
- Radiation Oncology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Morena Caliandro
- Radiation Oncology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | | | - Nicola Sasso
- Medical Oncology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Roberto Calbi
- Radiology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Maria Annunziata Gentile
- Radiology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Tiziana Talienti
- Radiation Oncology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Isabella Bruno
- Nuclear Medicine Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Michele Troia
- Pathology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Ilaria Bonaparte
- Radiation Oncology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Giuseppe Mario Ludovico
- Urology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Giammarco Surico
- Medical Oncology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
| | - Alba Fiorentino
- Radiation Oncology Department, General Regional Hospital F. Miulli, 70021 Acquaviva delle Fonti, BA, Italy
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, BA, Italy
| |
Collapse
|
6
|
Zheng J, Zheng Z, Zhang T, Chen X, Pang Q, Wang P, Yan C, Zhang W. Optimization of radiation target volume for locally advanced esophageal cancer in the immunotherapy era. Expert Opin Biol Ther 2024; 24:1221-1232. [PMID: 39460561 DOI: 10.1080/14712598.2024.2423009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 10/28/2024]
Abstract
INTRODUCTION Locally advanced esophageal cancer (EC) has poor prognosis. Preliminary clinical studies have demonstrated the synergistic efficacy of radiotherapy combined with immunotherapy in EC. Adjusting the radiotherapy target volume to protect immune function favors immunotherapy. However, there is no clear consensus on the exact definition of the EC target volume. AREAS COVERED Preclinical studies have provided a wealth of information on immunotherapy combined with different radiotherapy modalities, and several clinical studies have evaluated the impact of immunotherapy combined with radiotherapy on locally advanced EC. Here, we illustrate the rational target volume delineation for radiotherapy in terms of patient prognosis, pattern of radiotherapy failure, treatment-related toxicities, tumor-draining lymph nodes, and systemic immunity and summarize the clinical trials of radiotherapy combined with immunotherapy in EC. EXPERT OPINION We recommend applying involved-field irradiation (IFI) instead of elective nodal irradiation (ENI) for irradiated fields when immunotherapy is combined with chemoradiotherapy (CRT) for locally advanced EC. We expect that this target design will be evaluated in clinical trials to further explore more precise diagnostic modalities, long-term toxic responses, and quality of survival, and stratification factors for personalized treatment, and to provide more treatment benefits for patients.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhunhao Zheng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tian Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xi Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qingsong Pang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
7
|
Li X, Liu Y, Gui J, Gan L, Xue J. Cell Identity and Spatial Distribution of PD-1/PD-L1 Blockade Responders. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400702. [PMID: 39248327 PMCID: PMC11538707 DOI: 10.1002/advs.202400702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/08/2024] [Indexed: 09/10/2024]
Abstract
The programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) axis inhibits T cell activity, impairing anti-tumor immunity. Blocking this axis with therapeutic antibodies is one of the most promising anti-tumor immunotherapies. It has long been recognized that PD-1/PD-L1 blockade reinvigorates exhausted T (TEX) cells already present in the tumor microenvironment (TME). However, recent advancements in high-throughput gene sequencing and bioinformatic tools have provided researchers with a more granular and dynamic insight into PD-1/PD-L1 blockade-responding cells, extending beyond the TME and TEX populations. This review provides an update on the cell identity, spatial distribution, and treatment-induced spatiotemporal dynamics of PD-1/PD-L1 blockade responders. It also provides a synopsis of preliminary reports of potential PD-1/PD-L1 blockade responders other than T cells to depict a panoramic picture. Important questions to answer in further studies and the translational and clinical potential of the evolving understandings are also discussed.
Collapse
Affiliation(s)
- Xintong Li
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Yuanxin Liu
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Jun Gui
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Lu Gan
- Research Laboratory of Emergency MedicineDepartment of Emergency MedicineNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsLaboratory of Clinical Cell TherapyWest China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
8
|
Lei PJ, Fraser C, Jones D, Ubellacker JM, Padera TP. Lymphatic system regulation of anti-cancer immunity and metastasis. Front Immunol 2024; 15:1449291. [PMID: 39211044 PMCID: PMC11357954 DOI: 10.3389/fimmu.2024.1449291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer dissemination to lymph nodes (LN) is associated with a worse prognosis, increased incidence of distant metastases and reduced response to therapy. The LN microenvironment puts selective pressure on cancer cells, creating cells that can survive in LN as well as providing survival advantages for distant metastatic spread. Additionally, the presence of cancer cells leads to an immunosuppressive LN microenvironment, favoring the evasion of anti-cancer immune surveillance. However, recent studies have also characterized previously unrecognized roles for tumor-draining lymph nodes (TDLNs) in cancer immunotherapy response, including acting as a reservoir for pre-exhausted CD8+ T cells and stem-like CD8+ T cells. In this review, we will discuss the spread of cancer cells through the lymphatic system, the roles of TDLNs in metastasis and anti-cancer immune responses, and the therapeutic opportunities and challenges in targeting LN metastasis.
Collapse
Affiliation(s)
- Pin-Ji Lei
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Cameron Fraser
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Dennis Jones
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Jessalyn M. Ubellacker
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Timothy P. Padera
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
9
|
Tang X, Mao X, Ling P, Yu M, Pan H, Wang J, Liu M, Pan H, Qiu W, Che N, Zhang K, Bao F, Peng H, Ding Q, Wang S, Zhou W. Glycolysis inhibition induces anti-tumor central memory CD8 +T cell differentiation upon combination with microwave ablation therapy. Nat Commun 2024; 15:4665. [PMID: 38821965 PMCID: PMC11143264 DOI: 10.1038/s41467-024-49059-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/21/2024] [Indexed: 06/02/2024] Open
Abstract
Minimally invasive thermal therapy is a successful alternative treatment to surgery in solid tumors with high complete ablation rates, however, tumor recurrence remains a concern. Central memory CD8+ T cells (TCM) play important roles in protection from chronic infection and cancer. Here we find, by single-cell RNA analysis of human breast cancer samples, that although the memory phenotype of peripheral CD8+ T cells increases slightly after microwave ablation (MWA), the metabolism of peripheral CD8+ T cells remains unfavorable for memory phenotype. In mouse models, glycolysis inhibition by 2-deoxy-D-glucose (2DG) in combination with MWA results in long-term anti-tumor effect via enhancing differentiation of tumor-specific CD44hiCD62L+CD8+ TCM cells. Enhancement of CD8+ TCM cell differentiation determined by Stat-1, is dependent on the tumor-draining lymph nodes (TDLN) but takes place in peripheral blood, with metabolic remodeling of CD8+ T cells lasting the entire course of the the combination therapy. Importantly, in-vitro glycolysis inhibition in peripheral CD8+ T cells of patients with breast or liver tumors having been treated with MWA thrice leads to their differentiation into CD8+ TCM cells. Our work thus offers a potential strategy to avoid tumor recurrence following MWA therapy and lays down the proof-of-principle for future clinical trials.
Collapse
Affiliation(s)
- Xinyu Tang
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xinrui Mao
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Peiwen Ling
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Muxin Yu
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Hua Pan
- Department of General Surgery, Liyang Branch of Jiangsu Provincial People's Hospital, 70 Jianshe West Road, 213399, Liyang, China
| | - Jiaming Wang
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Mingduo Liu
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Hong Pan
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Wen Qiu
- Department of Immunology, Nanjing Medical University, Nanjing, 211166, China
| | - Nan Che
- Department of Rheumatology and Immunology, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China
| | - Kai Zhang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Pancreatic Center & Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
- Pancreas Institute of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Feifan Bao
- The First Clinical Medical College of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Hongwei Peng
- Department of General Surgery, Liyang Branch of Jiangsu Provincial People's Hospital, 70 Jianshe West Road, 213399, Liyang, China
| | - Qiang Ding
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Shui Wang
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Wenbin Zhou
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
10
|
Bao Y, Pan Z, Zhao L, Qiu J, Cheng J, Liu L, Qian D. BIBR1532 combined with radiotherapy induces ferroptosis in NSCLC cells and activates cGAS-STING pathway to promote anti-tumor immunity. J Transl Med 2024; 22:519. [PMID: 38816831 PMCID: PMC11138045 DOI: 10.1186/s12967-024-05331-3] [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: 12/08/2023] [Accepted: 05/18/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Telomerase, by safeguarding damaged telomeres and bolstering DNA damage repair, has the capacity to heighten the radioresistance of tumour cells. Thus, in turn, can compromise the efficacy of radiotherapy (RT) and radioimmunotherapy. Our previous studies have revealed that the highly selective telomerase inhibitor, BIBR1532, possesses the potential to enhance the radiosensitivity of Non-small cell lung cancer (NSCLC). In this study, we delve further into the impact of BIBR1532 on the immune activation induced by RT and elucidate the underlying mechanisms. METHODS Biological information analyses, immunofluorescence assays, western blot assays, flow cytometry analysis were conducted to elucidate the functions of the combination of BIBR1532 with radiotherapy in NSCLC. Intracellular levels of lipid peroxides, glutathione, malondialdehyde, and Fe2+ were measured as indicators of ferroptosis status. Both in vitro and in vivo studies were conducted to examine the antitumor effects. RESULTS Our findings indicate that the confluence of BIBR1532 with RT significantly augments the activation of the cGAS-STING pathway in both in vivo and in vitro settings, thereby fostering an effective anti-tumoral immune response. The effects can be ascribed to two key processes. Firstly, ionizing radiation, in precipitating DNA double-strand breaks (DSBs), prompts the release of tumour-derived double-stranded DNA (dsDNA) into the cytoplasm. Subsequently, BIBR1532 amplifies the activation of antigen-presenting cells by dsDNA post-RT and instigates the cGAS-STING pathway. Secondly, BIBR1532 enhances the ferroptosis response in NSCLC following RT, thereby promoting unrestrained lipid peroxidation and elevated levels of reactive oxygen species (ROS) within tumour cells. This ultimately leads to mitochondrial stress and the release of endogenous mitochondrial DNA (mtDNA) into the cytoplasm, thus facilitating the activation of the STING pathway and the induction of a type I interferon (IFN)-linked adaptive immune response. CONCLUSION This study underscores the potential of BIBR1532 as an efficacious and safe radiosensitizer and radioimmunotherapy synergist, providing robust preclinical research evidence for the treatment of NSCLC.
Collapse
Affiliation(s)
- Yawei Bao
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhipeng Pan
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, China
| | - Luqi Zhao
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204-5039, USA
| | - Jieping Qiu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jingjing Cheng
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Lei Liu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Dong Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
11
|
Yu T, Liu Z, Tao Q, Xu X, Li X, Li Y, Chen M, Liu R, Chen D, Wu M, Yu J. Targeting tumor-intrinsic SLC16A3 to enhance anti-PD-1 efficacy via tumor immune microenvironment reprogramming. Cancer Lett 2024; 589:216824. [PMID: 38522774 DOI: 10.1016/j.canlet.2024.216824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024]
Abstract
Immunotherapy, especially immune checkpoint inhibitors, has revolutionized clinical practice within the last decade. However, primary and secondary resistance to immunotherapy is common in patients with diverse types of cancer. It is well-acknowledged that tumor cells can facilitate the formation of immunosuppressive microenvironments via metabolism reprogramming, and lactic acid, the metabolite of glycolysis, is a significant contributor. SLC16A3 (also named as MCT4) is a transporter mediating lactic acid efflux. In this study, we investigated the role of glycolysis in immunotherapy resistance and aimed to improve the immunotherapy effects via Slc16a3 inhibition. Bioinformatical analysis revealed that the expression of glycolysis-related genes correlated with less CD8+ T cell infiltration and increased myeloid-derived suppressor cells (MDSC) enrichment. We found that high glycolytic activity in tumor cells adversely affected the antitumor immune responses and efficacy of immunotherapy and radiotherapy. As the transporter of lactic acid, SLC16A3 is highly expressed in glycolytic B16-F10 (RRID: CVCL_0159) cells, as well as human non-small cell lung carcinoma. We validated that Slc16a3 expression in tumor cells negatively correlated with anti-PD-1 efficiency. Overexpression of Slc16a3 in tumor cells promoted lactic acid production and efflux, and reduced tumor response to anti-PD-1 inhibitors by inhibiting CD8+ T cell function. Genetic and pharmacological inhibition of Slc16a3 dramatically reduced the glycolytic activity and lactic acid production in tumor cells, and ameliorated the immunosuppressive tumor microenvironments (TMEs), leading to boosted antitumor effects via anti-PD-1 blockade. Our study therefore demonstrates that tumor cell-intrinsic SLC16A3 may be a potential target to reverse tumor resistance to immunotherapy.
Collapse
Affiliation(s)
- Ting Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, PR China; Department of Oncology, Shandong University Cancer Center, Jinan, 250117, Shandong, PR China; Tianjin Medical University Cancer Institute &Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China; Cancer Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, PR China
| | - Zhaoyun Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, PR China; Department of Oncology, Shandong University Cancer Center, Jinan, 250117, Shandong, PR China
| | - Qingxu Tao
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, PR China
| | - Xin Xu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, PR China; Tianjin Medical University Cancer Institute &Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Xinyang Li
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, PR China; School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, PR China
| | - Yang Li
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, PR China; Department of Oncology, Shandong University Cancer Center, Jinan, 250117, Shandong, PR China
| | - Minxin Chen
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, PR China; Department of Oncology, Shandong University Cancer Center, Jinan, 250117, Shandong, PR China
| | - Rufei Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, PR China; Department of Oncology, Shandong University Cancer Center, Jinan, 250117, Shandong, PR China
| | - Dawei Chen
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, PR China; Department of Oncology, Shandong University Cancer Center, Jinan, 250117, Shandong, PR China
| | - Meng Wu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, PR China; Department of Oncology, Shandong University Cancer Center, Jinan, 250117, Shandong, PR China.
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, PR China; Department of Oncology, Shandong University Cancer Center, Jinan, 250117, Shandong, PR China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, PR China.
| |
Collapse
|
12
|
Liu Y, Jiang X, Wu Y, Yu H. Global research landscape and trends of cancer radiotherapy plus immunotherapy: A bibliometric analysis. Heliyon 2024; 10:e27103. [PMID: 38449655 PMCID: PMC10915415 DOI: 10.1016/j.heliyon.2024.e27103] [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: 09/03/2023] [Revised: 01/04/2024] [Accepted: 02/23/2024] [Indexed: 03/08/2024] Open
Abstract
The aim of this study was to present current research trends on the synergistic use of radiotherapy and immunotherapy (IRT) for cancer treatment. On March 1, 2023, we conducted a literature search for IRT papers using the Web of Science database. We extracted information and constructed two databases - the Core Database (CD) with 864 papers and Generalized Database (GD) with 6344 papers. A bibliometric analysis was performed to provide insights into the research landscape, to identify emerging trends and highly cited papers and journals in the field of IRT. The CD contained 864 papers that were collectively cited 31,818 times. Prominent journals in this area included the New England Journal of Medicine, Lancet Oncology, and the Journal of Clinical Oncology. Corresponding authors from the USA contributed the most publications. In recent years, lung cancer, melanoma, stereotactic radiotherapy, immune checkpoint inhibitors, and the tumor microenvironment emerged as hot research areas. This bibliometric analysis presented quantitative insights into research concerning IRT and proposed potential avenues for further exploration. Moreover, researchers can use our findings to select appropriate journals for publication or identify prospective collaborators. In summary, this bibliometric analysis provides a comprehensive overview of the historical progression and recent advancements in IRT research that may serve as inspiration for future investigations.
Collapse
Affiliation(s)
- Yanhao Liu
- School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, China
| | - Xu Jiang
- Department of Nuclear Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, China
| | - Yujuan Wu
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, China
| | - Haiming Yu
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, China
| |
Collapse
|
13
|
Salgado MTSF, Silva MCS, Fratelli C, Braga ARC, Lopes TBG, Ferreira E, da Silva ILD, Paiva LSD, Votto APDS. Bioactive C-phycocyanin exerts immunomodulatory and antitumor activity in mice with induced melanoma. Toxicol Appl Pharmacol 2024; 484:116874. [PMID: 38428464 DOI: 10.1016/j.taap.2024.116874] [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: 12/19/2023] [Revised: 02/09/2024] [Accepted: 02/24/2024] [Indexed: 03/03/2024]
Abstract
Melanoma is the most aggressive and deadly skin cancer. The difficulty in its treatment arises from its ability to suppress the immune system, making it crucial to find a substance that increases anti-tumor immunity. C-phycocyanin (C-PC) appears as a promising bioactive, with multifaceted effects against several cancers, but its efficacy against melanoma has only been tested in vitro. Therefore, we investigated C-PC's the anti-tumor and immunomodulatory action in a murine melanoma model. The tumor was subcutaneously induced in C57BL/6 mice by injecting B16F10 cells. The animals were injected subcutaneously with C-PC for three consecutive days. After euthanasia, the tumor was weighed and measured. The inguinal lymph node was removed, and the cells were stained with antibodies and analyzed by flow cytometry. The heart, brain and lung were analyzed by histopathology. C-PC increased the B cell population of the inguinal lymph node in percentage and absolute number. The absolute number of T lymphocytes and myeloid cells were also increased in the groups treated with C-PC. Thus, C-PC showed a positive immunomodulatory effect both animals with and without tumor. However, this effect was more pronounced in the presence of the tumor. Positive immune system modulation may be associated with a reduction in tumor growth in animals treated with C-PC. Administration of C-PC subcutaneously did not cause organ damage. Our findings demonstrate C-PC's immunomodulatory and anti-melanoma action, paving the way for clinical research with this bioactive.
Collapse
Affiliation(s)
- Mariana Teixeira Santos Figueiredo Salgado
- Programa de Pós-Graduação em Ciências Fisiológicas, ICB, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil; Laboratório de Cultura Celular, ICB, FURG, Rio Grande, RS, Brazil.
| | - Mayara Cristini Sebastião Silva
- Laboratório de Imunorregulação, Departamento de Imunobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Programa de Pós-Graduação em Patologia, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Camilly Fratelli
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Vila Mathias, Santos, SP, Brazil
| | | | | | - Enio Ferreira
- Laboratório do Comportamento Celular, ICB, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Istefani Luciene Dayse da Silva
- Programa de Pós-Graduação em Patologia, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, RJ, Brazil; Departamento de Patologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Luciana Souza de Paiva
- Laboratório de Imunorregulação, Departamento de Imunobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Programa de Pós-Graduação em Patologia, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Ana Paula de Souza Votto
- Programa de Pós-Graduação em Ciências Fisiológicas, ICB, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil; Laboratório de Cultura Celular, ICB, FURG, Rio Grande, RS, Brazil
| |
Collapse
|
14
|
Wang Y, Zhu T, Shi Q, Zhu G, Zhu S, Hou F. Tumor-draining lymph nodes: opportunities, challenges, and future directions in colorectal cancer immunotherapy. J Immunother Cancer 2024; 12:e008026. [PMID: 38242718 PMCID: PMC10806546 DOI: 10.1136/jitc-2023-008026] [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] [Accepted: 01/07/2024] [Indexed: 01/21/2024] Open
Abstract
Tumor-draining lymph nodes (TDLNs) are potential immunotherapy targets that could expand the population of patients with colorectal cancer (CRC) who may benefit from immunotherapy. Currently, pathological detection of tumor cell infiltration limits the acquisition of immune information related to the resected lymph nodes. Understanding the immune function and metastatic risk of specific stages of lymph nodes can facilitate better discussions on the removal or preservation of lymph nodes, as well as the timing of immunotherapy. This review summarized the contribution of TDLNs to CRC responses to immune checkpoint blockade therapy, local immunotherapy, adoptive cell therapy, and cancer vaccines, and discussed the significance of these findings for the development of diagnostics based on TDLNs and the potential implications for guiding immunotherapy after a definitive diagnosis. Molecular pathology and immune spectrum diagnosis of TDLNs will promote significant advances in the selection of immunotherapy options and predicting treatment efficacy.
Collapse
Affiliation(s)
- Yao Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tingting Zhu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Shi
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guanghui Zhu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Siwei Zhu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fenggang Hou
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Yip PL, You R, Chen MY, Chua MLK. Embracing Personalized Strategies in Radiotherapy for Nasopharyngeal Carcinoma: Beyond the Conventional Bounds of Fields and Borders. Cancers (Basel) 2024; 16:383. [PMID: 38254872 PMCID: PMC10814653 DOI: 10.3390/cancers16020383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Radiotherapy is the primary treatment modality for non-metastatic nasopharyngeal carcinoma (NPC) across all TN-stages. Locoregional control rates have been impressive even from the 2D radiotherapy (RT) era, except when the ability to deliver optimal dose coverage to the tumor is compromised. However, short- and long-term complications following head and neck RT are potentially debilitating, and thus, there has been much research investigating technological advances in RT delivery over the past decades, with the primary goal of limiting normal tissue damage. On this note, with a plateau in gains of therapeutic ratio by modern RT techniques, future advances have to be focused on individualization of RT, both in terms of dose prescription and the delineation of target volumes. In this review, we analyzed the guidelines and evidence related to contouring methods, and dose prescription for early and locoregionally advanced (LA-) NPC. Next, with the preference for induction chemotherapy (IC) in patients with LA-NPC, we assessed the evidence concerning radiotherapy adaptations guided by IC response, as well as functional imaging and contour changes during treatment. Finally, we discussed on RT individualization that is guided by EBV DNA assessment, and its importance in the era of combinatorial immune checkpoint blockade therapy with RT.
Collapse
Affiliation(s)
- Pui Lam Yip
- Department of Radiation Oncology, National University Cancer Institute, National University Hospital, Singapore 119074, Singapore;
| | - Rui You
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.Y.); (M.-Y.C.)
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
| | - Ming-Yuan Chen
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.Y.); (M.-Y.C.)
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Cooperative Surgical Ward of Nasopharyngeal Carcinoma, Faifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510700, China
| | - Melvin L. K. Chua
- Division of Medical Sciences, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore 168583, Singapore
- Division of Radiation Oncology, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore 168583, Singapore
- Oncology Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
16
|
Swamy K. Therapeutic In Situ Cancer Vaccine Using Pulsed Stereotactic Body Radiotherapy-A Translational Model. Vaccines (Basel) 2023; 12:7. [PMID: 38276666 PMCID: PMC10819354 DOI: 10.3390/vaccines12010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Both radiation and cancer therapeutic vaccine research are more than 100 years old, and their potential is likely underexplored. Antiangiogenics, nanoparticle targeting, and immune modulators are some other established anticancer therapies. In the meantime, immunotherapy usage is gaining momentum in clinical applications. This article proposes the concept of a pulsed/intermittent/cyclical endothelial-sparing single-dose in situ vaccination (ISVRT) schedule distinguishable from the standard therapeutic stereotactic body radiotherapy (SBRT) and stereotactic radiosurgery (SRS) plans. This ISVRT schedule can repeatedly generate tumor-specific neoantigens and epitopes for primary and immune modulation effects, augment supplementary immune enhancement techniques, activate long-term memory cells, avoid extracellular matrix fibrosis, and essentially synchronize with the vascular normalized immunity cycle. The core mechanisms of ISVRT impacting in situ vaccination would be optimizing cascading antigenicity and adjuvanticity. The present proposed hypothesis can be validated using the algorithm presented. The indications for the proposed concept are locally progressing/metastatic cancers that have failed standard therapies. Immunotherapy/targeted therapy, chemotherapy, antiangiogenics, and vascular-lymphatic normalization are integral to such an approach.
Collapse
|
17
|
Yokota T, Zenda S, Kodaira T, Kiyota N, Fujimoto Y, Wasano K, Takahashi R, Mizowaki T, Homma A, Sasaki K, Machida R, Sekino Y, Fukuda H. Novel approach of prophylactic radiation to reduce toxicities comparing 2-step40 with 56-Gy simultaneous integrated boost intensity-modulated radiation therapy for locally advanced squamous cell carcinoma of the head and neck, an intergroup phase III trial (JCOG1912, NEW BRIDGE). BMC Cancer 2023; 23:1068. [PMID: 37932681 PMCID: PMC10626703 DOI: 10.1186/s12885-023-11503-z] [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: 09/20/2023] [Accepted: 10/09/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Chemoradiotherapy (CRT) with concurrent cisplatin is the standard of care as a nonsurgical definitive treatment for patients with locally advanced squamous cell carcinoma of the head and neck (LA-SCCHN). However, CRT is associated with increased severe late adverse events, including swallowing dysfunction, xerostomia, ototoxicity, and hypothyroidism. Few strategies aimed at less invasive CRT without compromising treatment outcomes have been successful. The purpose of this study is to confirm the non-inferiority of reduced dose prophylactic radiation with 40 Gy compared to standard dose prophylactic radiation with 56 Gy in terms of the time to treatment failure (TTF) among patients with clinical stage III-IVB LA-SCCHN. METHODS This study is a multicenter, two-arm, open-label, randomized phase III trial. Patients with LA-SCCHN excluding p16 positive oropharynx cancer are randomized to the standard arm or experimental arm. A total dose of 70 Gy for tumors with concurrent cisplatin at 100 mg/m2 are administered in both arms. For prophylactic field, patients in the standard arm receive a total dose of 56 Gy in 35 fractions for 7 weeks using simultaneous integrated boost (SIB56) and those in the experimental arm receive 40 Gy in 20 fractions using two-step methods for 4 weeks (2-step40). A total of 400 patients will be enrolled from 52 Japanese institutions within 5 years. The primary endpoint is TTF, and the secondary endpoints are overall survival, complete response rate, progression-free survival, locoregional relapse-free survival, acute and late adverse events, quality of life score, and swallowing function score. DISCUSSION If the experimental arm is non-inferior to the standard arm in terms of TTF and superior on the safety endpoints, the 2-step40 procedure is the more useful treatment than SIB56 for definitive CRT. TRIAL REGISTRATION This trial has been registered in the Japan Registry of Clinical Trials as jRCTs031210100 ( https://jrct.niph.go.jp/latest-detail/jRCTs031210100 ). Date of Registration: May 2021.
Collapse
Affiliation(s)
- Tomoya Yokota
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Sunto-gun, Japan
| | - Sadamoto Zenda
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takeshi Kodaira
- Department of Radiation Oncology, Aichi Cancer Center Hospital, Nagoya, Japan, 1-1 Kanoko-den, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan.
| | - Naomi Kiyota
- Department of Medical Oncology and Hematology, Cancer Center, Kobe University Hospital, Kobe, Japan
| | - Yasushi Fujimoto
- Department of Otolaryngology, Aichi Medical University, Nagakute, Japan
| | - Koichiro Wasano
- Department of Otolaryngology-Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Ryo Takahashi
- Section of Radiation Safety and Quality Assurance, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takashi Mizowaki
- Departments of Radiation Oncology and Image-Applied Therapy, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akihiro Homma
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Keita Sasaki
- Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Ryunosuke Machida
- Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Yuta Sekino
- Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Haruhiko Fukuda
- Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
18
|
Wang J, Ge H, Tian Z. Immunotherapy Plus Radiotherapy for the Treatment of Sarcomas: Is There a Potential for Synergism? Onco Targets Ther 2023; 16:385-397. [PMID: 37313391 PMCID: PMC10258041 DOI: 10.2147/ott.s410693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Soft tissue sarcoma (STS) is a highly heterogeneous malignant tumor derived from mesenchymal tissue. Advanced STS has a poor response to the current anti-cancer therapeutic options, with a median overall survival of less than two years. Thus, new and more effective treatment methods for STS are needed. Increasing evidence has shown that immunotherapy and radiotherapy have synergistic therapeutic effects against malignant tumors. In addition, immunoradiotherapy has yielded positive results in clinical trials for various cancers. In this review, we discuss the synergistic mechanism of immunoradiotherapy in cancer treatment and the application of this combined regimen for the treatment of several cancers. In addition, we summarize the existing evidence on the use of immunoradiotherapy for the treatment of STS and the relevant clinical trials that are currently ongoing. Furthermore, we identify challenges in the use of immunoradiotherapy for the treatment of sarcomas and propose methods and precautions for overcoming these challenges. Lastly, we propose clinical research strategies and future research directions to help in the research and treatment of STS.
Collapse
Affiliation(s)
- Jiaqiang Wang
- Department of Bone and Soft Tissue, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan Province, 450008, People’s Republic of China
| | - Hong Ge
- Department of Radiotherapy, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan Province, 450008, People’s Republic of China
| | - Zhichao Tian
- Department of Bone and Soft Tissue, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan Province, 450008, People’s Republic of China
| |
Collapse
|
19
|
Zheng Y, Liu X, Li N, Zhao A, Sun Z, Wang M, Luo J. Radiotherapy combined with immunotherapy could improve the immune infiltration of melanoma in mice and enhance the abscopal effect. Radiat Oncol J 2023; 41:129-139. [PMID: 37403355 DOI: 10.3857/roj.2023.00185] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/08/2023] [Indexed: 07/06/2023] Open
Abstract
PURPOSE To analyze the gene mutation, immune infiltration and tumor growth of primary tumor and distant tumor under different treatment modes. MATERIALS AND METHODS Twenty B16 murine melanoma cells were injected subcutaneously into the of both sides of the thigh, simulating a primary tumor and a secondary tumor impacted by the abscopal effect, respectively. They were divided into blank control group, immunotherapy group, radiotherapy group, and radiotherapy combined immunotherapy group. During this period, tumor volume was measured, and RNA sequencing was performed on tumor samples after the test. R software was used to analyze differentially expressed genes, functional enrichment, and immune infiltration. RESULTS We found that any treatment mode could cause changes in differentially expressed genes, especially the combination treatment. The different therapeutic effects might be caused by gene expression. In addition, the proportions of infiltrating immune cells in the irradiated and abscopal tumors were different. In the combination treatment group, T-cell infiltration in the irradiated site was the most obvious. In the immunotherapy group, CD8+ T-cell infiltration in the abscopal tumor site was obvious, but immunotherapy alone might have a poor prognosis. Whether the irradiated or abscopal tumor was evaluated, radiotherapy combined with anti-programmed cell death protein 1 (anti-PD-1) therapy produced the most obvious tumor control and might have a positive impact on prognosis. CONCLUSION Combination therapy not only improves the immune microenvironment but may also have a positive impact on prognosis.
Collapse
Affiliation(s)
- Yufeng Zheng
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Xue Liu
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Department of Radiotherapy, Dalian Medical University, Dalian, China
| | - Na Li
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Aimei Zhao
- Department of Obstetrics and Gynecology, Liaocheng Dongchangfu District Maternal and Child Health Hospital, Liaocheng, China
| | - Zhiqiang Sun
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Meihua Wang
- Department of Pathology, Changzhou Fourth People's Hospital, Changzhou, China
| | - Judong Luo
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
20
|
Zhao J, Sun Y, Gao P, Zhao Z, Wei G. S-allylmercaptocysteine promotes anti-tumor immunity by suppressing PD-L1 expression. Biomed Pharmacother 2023; 161:114446. [PMID: 37002570 DOI: 10.1016/j.biopha.2023.114446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
SAMC (S-allylmercaptocysteine) possesses significant anti-tumor effects and is proven to inhibit inflammation in chronic obstructive pulmonary disease. The potential to regulate the immune system of SAMC inspired us to detect whether SAMC can promote anti-tumor immunity. Here we found that SAMC inhibits tumor development and progression by boosting CD8+ T cell and NK cell infiltration and decreasing the frequency of immune suppressing Treg cells in tumor tissue and enhancing the systemic immune function. Mechanistically, we found that SAMC suppresses PD-L1 expression at transcriptional level to increase the activation of anti-tumor cytotoxic T cells. Finally, we proved that SAMC inhibits PD-L1 transcription by suppressing the phosphorylation activation of STAT3. In conclusion, our findings reveal that SAMC is a potent immunity regulator and a potential agent for immune checkpoint inhibition in tumor therapy.
Collapse
|
21
|
Pierrard J, Van Ooteghem G, Van den Eynde M. Implications of the Organ-Specific Immune Environment for Immune Priming Effect of Radiotherapy in Metastatic Setting. Biomolecules 2023; 13:689. [PMID: 37189436 PMCID: PMC10136331 DOI: 10.3390/biom13040689] [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: 02/21/2023] [Revised: 04/07/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
With the development of immune checkpoint inhibitors (ICIs), the tumour immune microenvironment (TIME) has been increasingly considered to improve cancer management. The TIME of metastatic lesions is strongly influenced by the underlying immune contexture of the organ in which they are located. The metastatic location itself appears to be an important prognostic factor in predicting outcomes after ICI treatment in cancer patients. Patients with liver metastases are less likely to respond to ICIs than patients with metastases in other organs, likely due to variations in the metastatic TIME. Combining additional treatment modalities is an option to overcome this resistance. Radiotherapy (RT) and ICIs have been investigated together as an option to treat various metastatic cancers. RT can induce a local and systemic immune reaction, which can promote the patient's response to ICIs. Here, we review the differential impact of the TIME according to metastatic location. We also explore how RT-induced TIME modifications could be modulated to improve outcomes of RT-ICI combinations.
Collapse
Affiliation(s)
- Julien Pierrard
- UCLouvain, Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institute de Recherche Experimentale et Clinique (IREC), 1200 Brussels, Belgium
- Radiation Oncology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Geneviève Van Ooteghem
- UCLouvain, Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institute de Recherche Experimentale et Clinique (IREC), 1200 Brussels, Belgium
- Radiation Oncology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Marc Van den Eynde
- UCLouvain, Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institute de Recherche Experimentale et Clinique (IREC), 1200 Brussels, Belgium
- Medical Oncology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|