1
|
Pham TN, Coupey J, Yger F, Candéias SM, Thariat J, Valable S. Effect of glioblastoma and brain radiotherapy on T-lymphocyte subpopulations in rodents. Radiother Oncol 2025; 206:110801. [PMID: 40081500 DOI: 10.1016/j.radonc.2025.110801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/16/2025]
Abstract
INTRODUCTION Although lymphopenia is linked to immune suppression favoring tumor growth, the effect of different radiation types on specific T-lymphocyte subsets remains unclear. Among the T-lymphocyte subpopulations, CD8+-lymphocytes serve as key effectors in cancer immunity. This study aimed to examine the changes in T-lymphocyte subpopulations in both tumor-free and glioblastoma-bearing mice following brain-irradiation. METHODS The study was divided into two main phases. First, C57BL/6 mice, with or without glioblastoma (GL261 cells), received hemispheric brain-irradiation or no-treatment. T-lymphocyte subpopulations were analyzed using flow cytometry at various timepoint. The effect of tumor size and brain-irradiation on these cells was assessed using correlation analysis. Next, C57BL/6 mice were subjected to different brain-irradiation conditions. Blood samples were collected during and post-irradiation to analyze T-lymphocyte subpopulations, and tree-based modeling was used to determine radiation parameters impact on naïve CD8+-lymphocyte levels. RESULTS Glioblastoma reduced all T-lymphocyte subpopulations by day 15 post-inoculation. Radiotherapy decreased regulatory and effector CD4+-lymphocytes in both tumor-free and glioblastoma-bearing mice, but not naïve or memory CD4+-lymphocytes, in both tumor-free and glioblastoma-bearing mice. In tumor-free mice, radiotherapy had no effect on CD8+-lymphocytes, but reduced all CD8+-lymphocyte types in glioblastoma-bearing mice. Glioblastoma size negatively affected CD8+-lymphocytes. Brain-irradiation persistently reduced naïve and memory CD8+-lymphocytes, but effector and regulatory T-lymphocytes recovered. Exposure of lymph nodes to radiation worsened CD8+-lymphocyte reduction. CONCLUSION These findings confirm that the presence of glioblastoma and brain-irradiation affect T-lymphocyte subpopulations in mice. The inclusion of lymph nodes in the irradiated area led to a long-term decrease in naïve CD8+-lymphocytes. Further mechanistic studies are needed to understand the molecular basis of radiation impact on T-lymphocyte subpopulations.
Collapse
Affiliation(s)
- Thao-Nguyen Pham
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000 Caen, France; Laboratoire de Physique Corpusculaire UMR6534 IN2P3/ENSICAEN, France - Normandie Université, France
| | - Julie Coupey
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000 Caen, France
| | - Florian Yger
- Univ. Paris-Dauphine, PSL Research Univ./CNRS, LAMSADE, Paris, France
| | - Serge M Candéias
- Univ. Grenoble Alpes, CEA, CNRS, IRIG-LCBM-UMR5249, 38054 Grenoble, France
| | - Juliette Thariat
- Laboratoire de Physique Corpusculaire UMR6534 IN2P3/ENSICAEN, France - Normandie Université, France; Department of Radiation Oncology, Centre François Baclesse, Caen, Normandy, France.
| | - Samuel Valable
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000 Caen, France.
| |
Collapse
|
2
|
Jiang Q, Chen Z, Jiang J, Chen Q, Lan H, Zhu J, Mao W. The role of cGAS-STING in remodeling the tumor immune microenvironment induced by radiotherapy. Crit Rev Oncol Hematol 2025; 209:104658. [PMID: 39956501 DOI: 10.1016/j.critrevonc.2025.104658] [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: 11/04/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025] Open
Abstract
The activation of the cGAS-STING pathway occurs when tumor cell DNA is damaged by ionizing radiation. Once triggered, this pathway reshapes the tumor immune microenvironment by promoting the maturation, activation, polarization, and immune-killing capacity of immune cells, as well as by inducing the release of interferons and the expression of immune-related genes. In addition, the gut microbiota and various mechanisms of programmed cell death interact with the cGAS-STING pathway, further influencing its function in remodeling the immune microenvironment after radiotherapy. Therefore, investigating the mechanisms of the cGAS-STING pathway in reshaping the tumor immune microenvironment post-radiotherapy can not only optimize the efficacy of combined radiotherapy and immunotherapy but also provide new research directions and potential targets for cancer treatment.
Collapse
Affiliation(s)
- Qingyu Jiang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Zhejiang Chinese Medical University, Hangzhou 310053, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Zhiheng Chen
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing 31400, China
| | - Jin Jiang
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing 31400, China
| | - Qianping Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Huiyin Lan
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Ji Zhu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China.
| | - Wei Mao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China.
| |
Collapse
|
3
|
Zheng L, Cai W, Ke Y, Hu X, Yang C, Zhang R, Wu H, Liu D, Yu H, Wu C. Cancer‑associated fibroblasts: a pivotal regulator of tumor microenvironment in the context of radiotherapy. Cell Commun Signal 2025; 23:147. [PMID: 40114180 PMCID: PMC11927177 DOI: 10.1186/s12964-025-02138-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND In the course of tumor treatment, radiation therapy (RT) not only kills cancer cells, but also induces complex biological effects in non-malignant cells around cancer cells. These biological effects such as angiogenesis, changes in stromal composition and immune cell infiltration remodel the tumor microenvironment (TME). As one of the major components of the TME, Cancer‑associated fibroblasts (CAFs) are not only involved in tumorigenesis, progression, recurrence, and metastasis but also regulate the tumor-associated immune microenvironment. CAFs and tumor cells or immune cells have complex intercellular communication in the context of tumor radiation. MAIN CONTENT Different cellular precursors, spatial location differences, absence of specific markers, and advances in single-cell sequencing technology have gradually made the abundant heterogeneity of CAFs well known. Due to unique radioresistance properties, CAFs can survive under high doses of ionizing radiation. However, radiation can induce phenotypic and functional changes in CAFs and further act on tumor cells and immune cells to promote or inhibit tumor progression. To date, the effect of RT on CAFs and the effect of irradiated CAFs on tumor progression and TME are still not well defined. CONCLUSION In this review, we review the origin, phenotypic, and functional heterogeneity of CAFs and describe the effects of RT on CAFs, focusing on the mutual crosstalk between CAFs and tumor or immune cells after radiation. We also discuss emerging strategies for targeted CAFs therapy.
Collapse
Affiliation(s)
- Linhui Zheng
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Wenqi Cai
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Yuan Ke
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Xiaoyan Hu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Chunqian Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Runze Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Huachao Wu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Dong Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Haijun Yu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071, China.
| | - Chaoyan Wu
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
| |
Collapse
|
4
|
Zhou S, Yang H. Radiotherapy modulates autophagy to reshape the tumor immune microenvironment to enhance anti-tumor immunity in esophageal cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189302. [PMID: 40120778 DOI: 10.1016/j.bbcan.2025.189302] [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: 07/29/2024] [Revised: 03/15/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
The combination of radiotherapy and immunotherapy exerts synergistic antitumor in a range of human cancers, and also in esophageal cancer. Radiotherapy-induced tumor immune microenvironment (TIME) reprogramming is an essential basis for the synergistic antitumor between radiotherapy and immunotherapy. Radiotherapy can induce autophagy in tumor cells and immune cells of TIME, and autophagy activation is involved in the modification of immunological characteristics of TIME. The TIME landscape of esophageal cancer, especially ESCC, can be affected by radiotherapy or autophagy regulation. In this review, we depicted that local radiotherapy-induced autophagy could promote the maturation, migration, infiltration, and function of immune cells by complicated mechanisms to make TIME from immune "cold" to "hot", resulting in the synergistic antitumor of RT and IO. We argue that unraveling the relevance of radiotherapy-initiated autophagy to driving radiotherapy reprogramming TIME will open new ideas to explore new targets or more efficiently multimodal therapeutic interventions in ESCC.
Collapse
Affiliation(s)
- Suna Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Department of Radiation Oncology, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China
| | - Haihua Yang
- Key Laboratory of Radiation Oncology of Taizhou, Department of Radiation Oncology, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China.
| |
Collapse
|
5
|
Gao D, Zhang H, Sun W, Wang H, Wang H. Radiation-Induced Intestinal Injury: Molecular Mechanisms and Therapeutic Status. DNA Cell Biol 2024; 43:537-548. [PMID: 39235407 DOI: 10.1089/dna.2024.0105] [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] [Indexed: 09/06/2024] Open
Abstract
Radiation-induced intestinal injury is one of the most common intestinal complications caused by pelvic and abdominal tumor radiotherapy, severely impacting patients' quality of life. Ionizing radiation, while killing tumor cells, inevitably damages healthy tissue. Radiation-induced enteropathy results from radiation therapy-induced intestinal tissue damage and inflammatory responses. This damage involves various complex molecular mechanisms, including cell apoptosis, oxidative stress, release of inflammatory mediators, disruption of immune responses, and imbalance of intestinal microbiota. A thorough understanding of these molecular mechanisms is crucial for developing effective prevention and treatment strategies.
Collapse
Affiliation(s)
- Dandan Gao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, China
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin 300121, China
| | - Heng Zhang
- Department of Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, China
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin 300121, China
| | - Wanjun Sun
- Department of Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, China
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin 300121, China
| | - Huaqing Wang
- Department of Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, China
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin 300121, China
| | - Hui Wang
- Department of Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, China
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin 300121, China
| |
Collapse
|
6
|
Wu Y, Yi M, Niu M, Zhou B, Mei Q, Wu K. Beyond success: unveiling the hidden potential of radiotherapy and immunotherapy in solid tumors. Cancer Commun (Lond) 2024; 44:739-760. [PMID: 38837878 PMCID: PMC11260771 DOI: 10.1002/cac2.12576] [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: 12/12/2023] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
Immunotherapy, particularly with immune checkpoint inhibitors, has significantly transformed cancer treatment. Despite its success, many patients struggle to respond adequately or sustain long-lasting clinical improvement. A growing consensus has emerged that radiotherapy (RT) enhances the response rate and overall efficacy of immunotherapy. Although combining RT and immunotherapy has been extensively investigated in preclinical models and has shown promising results, establishing itself as a dynamic and thriving area of research, clinical evidence for this combination strategy over the past five years has shown both positive and disappointing results, suggesting the need for a more nuanced understanding. This review provides a balanced and updated analysis of the combination of immunotherapy and RT. We summarized the preclinical mechanisms through which RT boosts antitumor immune responses and mainly focused on the outcomes of recently updated clinical trials, including those that may not have met expectations. We investigated the optimization of the therapeutic potential of this combined strategy, including key challenges, such as fractionation and scheduling, lymph node irradiation, and toxicity. Finally, we offered insights into the prospects and challenges associated with the clinical translation of this combination therapy, providing a realistic perspective on the current state of research and potential future directions.
Collapse
Affiliation(s)
- Yuze Wu
- Department of OncologyTongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Ming Yi
- Department of Breast SurgeryZhejiang University School of Medicine First Affiliated HospitalHangzhouZhejiangP. R. China
| | - Mengke Niu
- Department of OncologyTongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Binghan Zhou
- Department of OncologyTongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Qi Mei
- Department of OncologyTongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Kongming Wu
- Cancer CenterShanxi Bethune HospitalShanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical UniversityTaiyuanShanxiP. R. China
- Cancer CenterTongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| |
Collapse
|
7
|
Jin Y, Jiang J, Mao W, Bai M, Chen Q, Zhu J. Treatment strategies and molecular mechanism of radiotherapy combined with immunotherapy in colorectal cancer. Cancer Lett 2024; 591:216858. [PMID: 38621460 DOI: 10.1016/j.canlet.2024.216858] [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: 09/17/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024]
Abstract
Radiotherapy (RT) remodels the tumor immune microenvironment (TIME) and modulates the immune response to indirectly destroy tumor cells, in addition to directly killing tumor cells. RT combined with immunotherapy may significantly enhance the efficacy of RT in colorectal cancer by modulating the microenvironment. However, the molecular mechanisms by which RT acts as an immunomodulator to modulate the immune microenvironment remain unclear. Further, the optimal modalities of RT combined with immunotherapy for the treatment of colorectal cancer, such as the time point of combining RT and immunization, the fractionation pattern and dosage of radiotherapy, and other methods to improve the efficacy, are also being explored parallelly. To address these aspects, in this review, we summarized the mechanisms by which RT modulates TIME and concluded the progress of RT combined with immunization in preclinical and clinical trials. Finally, we discussed heavy ion radiation therapy and the efficacy of prediction markers and other immune combination therapies. Overall, combining RT with immunotherapy to enhance antitumor effects will have a significant clinical implication and will help to facilitate individualized treatment modalities.
Collapse
Affiliation(s)
- Yuzhao Jin
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China; Wenzhou Medical University, Wenzhou, 325000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China
| | - Jin Jiang
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, 31400, China
| | - Wei Mao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China
| | - Minghua Bai
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China
| | - Qianping Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China.
| | - Ji Zhu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China; Wenzhou Medical University, Wenzhou, 325000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China.
| |
Collapse
|
8
|
Song D, Ding Y. A new target of radiotherapy combined with immunotherapy: regulatory T cells. Front Immunol 2024; 14:1330099. [PMID: 38259489 PMCID: PMC10800811 DOI: 10.3389/fimmu.2023.1330099] [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: 10/30/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Radiotherapy is one important treatment for malignant tumours. It is widely believed today that radiotherapy has not only been used as a local tumour treatment method, but also can induce systemic anti-tumour responses by influencing the tumour microenvironment, but its efficacy is limited by the tumour immunosuppression microenvironment. With the advancement of technology, immunotherapy has entered a golden age of rapid development, gradually occupying a place in clinical tumour treatment. Regulatory T cells (Tregs) widely distributing in the tumour microenvironment play an important role in mediating tumour development. This article analyzes immunotherapy, the interaction between Tregs, tumours and radiotherapy. It briefly introduces immunotherapies targeting Tregs, aiming to provide new strategies for radiotherapy combined with Immunotherapy.
Collapse
Affiliation(s)
| | - Yun Ding
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
9
|
Keisari Y, Kelson I. Tumor ablation induced anti-tumor immunity: destruction of the tumor in situ with the aim to evoke a robust anti-tumor immune response. Cancer Metastasis Rev 2023; 42:1065-1068. [PMID: 37952066 PMCID: PMC10713665 DOI: 10.1007/s10555-023-10150-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Affiliation(s)
- Yona Keisari
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.
| | - Itzhak Kelson
- Sackler Faculty of Exact Sciences, School of Physics and Astronomy, Tel Aviv University, 6997801, Tel Aviv, Israel
| |
Collapse
|
10
|
Wu Z, Lin Q, Sheng L, Chen W, Liang M, Wu D, Ke Y. A novel immune-related risk-scoring system associated with the prognosis and response of cervical cancer patients treated with radiation therapy. Front Mol Biosci 2023; 10:1297774. [PMID: 38028542 PMCID: PMC10667679 DOI: 10.3389/fmolb.2023.1297774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Objective: The tumor microenvironment plays a critical role in the radiotherapy and immunotherapy response of cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC). Radioresistance is a key factor in treatment failure among patients who receive radical radiotherapy. Thus, new immune-related biomarkers associated with radiotherapy response in CESC are needed. Methods: In this study, the CIBERSORT and ESTIMATE methods were applied to determine the percentage of tumor-infiltrating cells and the number of immune components in 103 CESCs treated with radiotherapy from The Cancer Genome Atlas (TCGA) database. The main dysregulated genes were subjected to multivariate and univariate analyses. The prognostic value of this system was studied via receiver operating characteristic curve and survival analysis. For further confirmation, the biomarkers' expression levels and predictive value were validated by immunohistochemistry (IHC) and qRT-PCR. The CIBERSORT algorithm was used to calculate the compositional patterns of 22 types of immune cells in cervical cancer patients treated with radiation therapy. Results: Data for 17 radioresistant and 86 radiosensitive tumors were obtained from the The Cancer Genome Atlas database. 53 immune-related DEGs were identified. GO and KEGG analyses revealed that the DEGs were enriched in protein kinase B signaling, growth factors in cytokines, the MAPK pathway and the PI3K-Akt pathway. Then, 14 key immune-related genes built a risk scoring model were deemed prognostic in CESC with radiotherapy. The area under the curve (AUC) of the model was 0.723, and the high-risk group presented worse outcomes than the low-risk group. In addition, the high-risk group tended to have persistent tumors (p = 0.001). The high expression of WT1 and SPOUYT4 were associated with relapse, the high expression of Angiotensinogen and MIEN1 were associated with nonrelapse. Analysis of the immune microenvironment indicated that M0 macrophages, M2 macrophages, activated mast cells and resting memory CD4+ T cells were positively correlated with the risk score (p < 0.05). Conclusion: The novel immune-related risk scoring system has some advantages in predicting the prognosis and treatment response of cervical cancer patients treated with radiotherapy. Moreover, it might provide novel clues for providing targeted immune therapy to these patients.
Collapse
Affiliation(s)
- Zhuna Wu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qiuya Lin
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Liying Sheng
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Weihong Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Meili Liang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Danni Wu
- Department of Operation, The Second Hospital of Jinjiang, Quanzhou, China
| | - Yumin Ke
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
11
|
Ding Y, Weng S, Zhu N, Mi M, Xu Z, Zhong L, Yuan Y. Immunotherapy combined with local therapy in the late-line treatment of repair-proficient (pMMR)/microsatellite stable (MSS) metastatic colorectal cancer. Heliyon 2023; 9:e22092. [PMID: 38058653 PMCID: PMC10695980 DOI: 10.1016/j.heliyon.2023.e22092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies, and at the initial visit, most patients are diagnosed with metastatic CRC (mCRC). However, immunotherapy is only and highly effective in a very small proportion of patients with mCRC having mismatch repair defect (dMMR)/high microsatellite instability, and the majority of the patients with mCRC having mismatch repair proficient (pMMR)/microsatellite stability (MSS) cannot benefit from it. At present, many clinical studies of immunotherapy combined with tyrosine kinase inhibitors (TKIs) are trying to regulate the immune microenvironment of pMMR/MSS mCRC, transforming a "cold tumor" into a "hot tumor," which has not only surprising effects but also certain limitations, i.e., the response could not be specific to metastasis. Therefore, regarding the bottleneck encountered by immunotherapy in patients with patients pMMR/MSS mCRC, this study summarized current research and possible mechanisms of immunotherapy combined with local therapy for metastasis, including radiotherapy, ablation, and transcatheter arterial chemoembolization.
Collapse
Affiliation(s)
- Yuwei Ding
- Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang Province, China
| | - Shanshan Weng
- Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ning Zhu
- Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang Province, China
| | - Mi Mi
- Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang Province, China
| | - Ziheng Xu
- Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang Province, China
| | - Liping Zhong
- Department of Oncology, Huzhou Central Hospital, Huzhou, Zhejiang Province, China
| | - Ying Yuan
- Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang Province, China
- Cancer Center of Zhejiang University, Hangzhou, Zhejiang Province, China
- Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, 310052, China
| |
Collapse
|
12
|
Zhao D, Mo Y, Neganova ME, Aleksandrova Y, Tse E, Chubarev VN, Fan R, Sukocheva OA, Liu J. Dual effects of radiotherapy on tumor microenvironment and its contribution towards the development of resistance to immunotherapy in gastrointestinal and thoracic cancers. Front Cell Dev Biol 2023; 11:1266537. [PMID: 37849740 PMCID: PMC10577389 DOI: 10.3389/fcell.2023.1266537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023] Open
Abstract
Successful clinical methods for tumor elimination include a combination of surgical resection, radiotherapy, and chemotherapy. Radiotherapy is one of the crucial components of the cancer treatment regimens which allow to extend patient life expectancy. Current cutting-edge radiotherapy research is focused on the identification of methods that should increase cancer cell sensitivity to radiation and activate anti-cancer immunity mechanisms. Radiation treatment activates various cells of the tumor microenvironment (TME) and impacts tumor growth, angiogenesis, and anti-cancer immunity. Radiotherapy was shown to regulate signaling and anti-cancer functions of various TME immune and vasculature cell components, including tumor-associated macrophages, dendritic cells, endothelial cells, cancer-associated fibroblasts (CAFs), natural killers, and other T cell subsets. Dual effects of radiation, including metastasis-promoting effects and activation of oxidative stress, have been detected, suggesting that radiotherapy triggers heterogeneous targets. In this review, we critically discuss the activation of TME and angiogenesis during radiotherapy which is used to strengthen the effects of novel immunotherapy. Intracellular, genetic, and epigenetic mechanisms of signaling and clinical manipulations of immune responses and oxidative stress by radiotherapy are accented. Current findings indicate that radiotherapy should be considered as a supporting instrument for immunotherapy to limit the cancer-promoting effects of TME. To increase cancer-free survival rates, it is recommended to combine personalized radiation therapy methods with TME-targeting drugs, including immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Deyao Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingyi Mo
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Margarita E. Neganova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russia
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Chernogolovka, Russia
| | - Yulia Aleksandrova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russia
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Chernogolovka, Russia
| | - Edmund Tse
- Department of Hepatology, Royal Adelaide Hospital, CALHN, Adelaide, SA, Australia
| | - Vladimir N. Chubarev
- Sechenov First Moscow State Medical University, Sechenov University, Moscow, Russia
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Olga A. Sukocheva
- Department of Hepatology, Royal Adelaide Hospital, CALHN, Adelaide, SA, Australia
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Liao W, Liu C, Yang K, Chen J, Wu Y, Zhang S, Yu K, Wang L, Ran L, Chen M, Chen F, Xu Y, Wang S, Wang F, Zhang Q, Zhao J, Ye L, Du C, Wang J. Aged hematopoietic stem cells entrap regulatory T cells to create a prosurvival microenvironment. Cell Mol Immunol 2023; 20:1216-1231. [PMID: 37644165 PMCID: PMC10541885 DOI: 10.1038/s41423-023-01072-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 07/02/2023] [Accepted: 07/19/2023] [Indexed: 08/31/2023] Open
Abstract
Although DNA mutation drives stem cell aging, how mutation-accumulated stem cells obtain clonal advantage during aging remains poorly understood. Here, using a mouse model of irradiation-induced premature aging and middle-aged mice, we show that DNA mutation accumulation in hematopoietic stem cells (HSCs) during aging upregulates their surface expression of major histocompatibility complex class II (MHCII). MHCII upregulation increases the chance for recognition by bone marrow (BM)-resident regulatory T cells (Tregs), resulting in their clonal expansion and accumulation in the HSC niche. On the basis of the establishment of connexin 43 (Cx43)-mediated gap junctions, BM Tregs transfer cyclic adenosine monophosphate (cAMP) to aged HSCs to diminish apoptotic priming and promote their survival via activation of protein kinase A (PKA) signaling. Importantly, targeting the HSC-Treg interaction or depleting Tregs effectively prevents the premature/physiological aging of HSCs. These findings show that aged HSCs use an active self-protective mechanism by entrapping local Tregs to construct a prosurvival niche and obtain a clonal advantage.
Collapse
Affiliation(s)
- Weinian Liao
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Chaonan Liu
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Ke Yang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, China
| | - Jun Chen
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Yiding Wu
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Shuzhen Zhang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Kuan Yu
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Lisha Wang
- Institute of Immunology, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Li Ran
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, China
| | - Mo Chen
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Fang Chen
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Yang Xu
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Song Wang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Fengchao Wang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Qian Zhang
- National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, 200433, Shanghai, China
| | - Jinghong Zhao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, China
| | - Lilin Ye
- Institute of Immunology, Army Medical University (Third Military Medical University), 400038, Chongqing, China.
| | - Changhong Du
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China.
| | - Junping Wang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), 400038, Chongqing, China.
| |
Collapse
|
14
|
Zhu DQ, Su C, Li JJ, Li AW, Luv Y, Fan Q. Update on Radiotherapy Changes of Nasopharyngeal Carcinoma Tumor Microenvironment. World J Oncol 2023; 14:350-357. [PMID: 37869238 PMCID: PMC10588496 DOI: 10.14740/wjon1645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/11/2023] [Indexed: 10/24/2023] Open
Abstract
The utilization of radiotherapy (RT) serves as the principal approach for managing nasopharyngeal carcinoma (NPC). Consequently, it is imperative to investigate the correlation between the radiation microenvironment and radiation resistance in NPC. PubMed and China National Knowledge Infrastructure (CNKI) databases were accessed to perform a search utilizing the English keywords "nasopharyngeal cancer", "radiotherapy", and "microenvironment". The search time spanned from the establishment of the database until January 20, 2023. A total of 82 articles were included. The post-radiation tumor microenvironment (TME), or the radiation microenvironment, includes several components, such as the radiation-immune microenvironment and the radiation-hypoxic microenvironment. The radiation-immune microenvironment includes various factors like immune cells, signaling molecules, and extracellular matrix. RT can reshape the TME, leading to immune responses with both cytotoxic effects (T cells, B cells, natural killer (NK) cells) and immune escape mechanisms (regulatory T cells (Tregs), macrophages). RT enhances immune responses through DNA release, type I interferons, and immune cell recruitment. Radiation-hypoxic microenvironment affects metabolism and molecular changes. RT-induced hypoxia causes vascular changes, fibrosis, and vessel compression, leading to tissue hypoxia. Hypoxia activates hypoxia-inducible factor (HIF)-1α/2α, promoting angiogenesis and glycolysis in tumor cells. TME changes due to hypoxia also involve immune suppressive cells like myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), and Tregs. The radiation microenvironment is involved in radiation resistance and holds a significant effect on the prognosis of patients with NPC. Exploring the radiation microenvironment provides new insights into RT and NPC research.
Collapse
Affiliation(s)
- Dao Qi Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Chao Su
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jing Jun Li
- NanFang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ai Wu Li
- NanFang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ying Luv
- NanFang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qin Fan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
15
|
Koga F. Selective multimodal bladder-sparing therapy for muscle-invasive bladder cancer: the present and the future. Expert Rev Anticancer Ther 2023; 23:1127-1139. [PMID: 37753554 DOI: 10.1080/14737140.2023.2257389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
INTRODUCTION Despite the lack of level 1 evidence, selective bladder-sparing therapy using trimodal therapy is currently recommended by guidelines as a standard of care in patients with non-metastatic, muscle-invasive bladder cancer who are eligible for the treatment. AREAS COVERED This article reviews major studies of selective, bladder-sparing therapy utilizing multiple modalities for muscle-invasive bladder cancer and those comparing the oncological outcomes between bladder-sparing therapy and radical cystectomy. Also discussed are predictive biomarkers potentially capable of guiding treatment decisions by patients with muscle-invasive bladder cancer and a novel strategy for boosting the antitumor immune response in bladder-sparing therapy. PubMed databases were searched for records of 30 June 2023 or earlier. EXPERT OPINION Selective, bladder-sparing therapy appears to be underutilized at present. To promote its use, measures should be taken to facilitate the referral of eligible patients to specialist centers and broaden the number of facilities providing the therapy. Recent studies have suggested a prognostic benefit of radiotherapy for the primary lesion in patients with metastatic bladder cancer. Given that irradiation can induce the abscopal effect, particularly in combination with immune checkpoint inhibitors, demand for bladder-sparing therapies may increase in the context of treatments for metastases.
Collapse
Affiliation(s)
- Fumitaka Koga
- Department of Urology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
16
|
Palermo B, Bottero M, Panetta M, Faiella A, Sperduti I, Masi S, Frisullo G, Foddai ML, Cordone I, Nisticò P, Sanguineti G. Stereotactic Ablative Radiation Therapy in 3 Fractions Induces a Favorable Systemic Immune Cell Profiling in Prostate Cancer Patients. Oncoimmunology 2023; 12:2174721. [PMID: 36798427 PMCID: PMC9928462 DOI: 10.1080/2162402x.2023.2174721] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
The impact of radiotherapy (RT) on immune cell status in prostate cancer (PCa) is only partially determined. The aim of this study was to assess the effect of different RT strategies on peripheral B, T, and Natural killer (NK) lymphocytes at precise longitudinal time-points in PCa. 18 patients treated with stereotactic body radiation therapy (SBRT) (40 Gy/3FRX), definitive moderate-hypofractionation (62 Gy/20FRX), or post-operative conventional-fractionation RT (66-69 Gy/30FRX) were prospectively evaluated for the immune cell profile in terms of immune cell composition, differentiation stage, cytokine production and inhibitory receptor (IR) expression. The immune-monitoring of the 18 patients revealed that RT affects the balance of systemic immune cells, with the main differences observed between SBRT and conventionally fractionated RT. SBRT favorably impacts immune response in term of increased B cells, central-memory and effector-memory CD8+ T cells, along with decreased Treg cells after treatment. On the contrary, conventional fractionated RT had a long-term negative effect on the systemic immune profile, including a decrease of total lymphocyte counts accompanied by an increase of neutrophils-to-lymphocytes ratio. Total B and T cells decreased and Treg-to-CD8+ ratio increased. Functionality of T lymphocytes were not affected by any of the 3-fractionation schedules. Interestingly, SBRT significantly up-regulates the expression of V-domain immunoglobulin suppressor of T-cell activation (VISTA) in CD8+ T cells in the absence of other IRs. Our results indicate the relevance of systematic immunomonitoring during RT to identify novel immune-related target to design trials of combined radio-immunotherapy as a promising strategy in the clinical management of PCa.
Collapse
Affiliation(s)
- Belinda Palermo
- Unit Tumor Immunology and Immunotherapy, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marta Bottero
- Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Mariangela Panetta
- Unit Tumor Immunology and Immunotherapy, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Adriana Faiella
- Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Isabella Sperduti
- Biostatistical Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Serena Masi
- Clinical Pathology and Cancer Biobank, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppe Frisullo
- Unit Tumor Immunology and Immunotherapy, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Laura Foddai
- Transfusion Medicine, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Iole Cordone
- Clinical Pathology and Cancer Biobank, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Nisticò
- Unit Tumor Immunology and Immunotherapy, IRCCS Regina Elena National Cancer Institute, Rome, Italy,Paola Nisticò Unit Tumor Immunology and Immunotherapy, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppe Sanguineti
- Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy,CONTACT Giuseppe Sanguineti
| |
Collapse
|
17
|
Chen L, Shi V, Wang S, Freeman R, Ruiz F, Jayachandran K, Zhang J, Cosper P, Sun L, Luke CJ, Spina C, Grigsby PW, Schwarz JK, Markovina S. SCCA1/SERPINB3 promotes suppressive immune environment via STAT-dependent chemokine production, blunting the therapy-induced T cell responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526675. [PMID: 36778224 PMCID: PMC9915608 DOI: 10.1101/2023.02.01.526675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Radiotherapy is a commonly used cancer treatment; however, patients with high serum squamous cell carcinoma antigen (SCCA1/SERPINB3) are associated with resistance and poor prognosis. Despite being a strong clinical biomarker, the modulation of SERPINB3 in tumor immunity is poorly understood. We investigated the microenvironment of SERPINB3 high tumors through RNAseq of primary cervix tumors and found that SERPINB3 was positively correlated with CXCL1/8, S100A8/A9 and myeloid cell infiltration. Induction of SERPINB3 in vitro resulted in increased CXCL1/8 and S100A8/A9 production, and supernatants from SERPINB3-expressing cultures attracted monocytes and MDSCs. In murine tumors, the orthologue mSerpinB3a promoted MDSC, TAM, and M2 macrophage infiltration contributing to an immunosuppressive phenotype, which was further augmented upon radiation. Radiation-enhanced T cell response was muted in SERPINB3 tumors, whereas Treg expansion was observed. A STAT-dependent mechanism was implicated, whereby inhibiting STAT signaling with ruxolitinib abrogated suppressive chemokine production. Patients with elevated pre-treatment serum SCCA and high pSTAT3 had increased intratumoral CD11b+ myeloid cell compared to patients with low SCCA and pSTAT3 cohort that had overall improved cancer specific survival after radiotherapy. These findings provide a preclinical rationale for targeting STAT signaling in tumors with high SERPINB3 to counteract the immunosuppressive microenvironment and improve response to radiation.
Collapse
|
18
|
The Current and Future Promises of Combination Radiation and Immunotherapy for Genitourinary Cancers. Cancers (Basel) 2022; 15:cancers15010127. [PMID: 36612124 PMCID: PMC9818005 DOI: 10.3390/cancers15010127] [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: 11/13/2022] [Revised: 12/12/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
As the indications for the use of immunotherapy in genitourinary malignancies expand, its role in combination with standard or conventional therapies has become the subject of contemporary studies. Radiotherapy has multiple immunomodulating effects on anti-tumor immune response, which highlights potential synergistic role with immunotherapy agents. We sought to review the body of published data studying the combination of immunotherapy and radiotherapy as well as the rationale for combination therapy. Trial information and primary articles were obtained using the following terms "immunotherapy", "radiotherapy", "prostate cancer", and "bladder cancer." All articles and trials were screened to ensure they included combination radiotherapy and immunotherapy. The effects of radiation on the immune system, including both immunogenic and immunosuppressive effects, have been reported. There is a potential for combinatorial or synergistic effects between radiation therapy and immunotherapy in treating bladder and prostate cancers. However, results from ongoing and future clinical trials are needed to best integrate immunotherapy into current standard of care treatments for GU cancers.
Collapse
|
19
|
Jiang M, Qi Y, Huang W, Lin Y, Li B. Curcumin Reprograms TAMs from a Protumor Phenotype towards an Antitumor Phenotype via Inhibiting MAO-A/STAT6 Pathway. Cells 2022; 11:3473. [PMID: 36359867 PMCID: PMC9655729 DOI: 10.3390/cells11213473] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/20/2022] [Accepted: 10/28/2022] [Indexed: 07/30/2023] Open
Abstract
M1 phenotype macrophages have anticancer characteristics, whereas M2 phenotype macrophages promote tumor growth and metastasis. A higher M1/M2 ratio, therefore, has a beneficial effect on the tumor immune microenvironment, thereby inhibiting tumor growth. The natural alkaloid curcumin is found to have anticancer properties. However, the mechanism remains unclear. In this study, a cell co-culture system and M2 macrophage model were used to evaluate the effects of curcumin on tumor-associated macrophage (TAM) phenotypes. Our results demonstrate that curcumin reprogrammed the M2 macrophages by reducing the level of anti-inflammatory cytokines (TGF-β, Arg-1, and IL-10) and an M2 surface marker (CD206) induced by Cal27 cells or IL-4, as well as upregulating proinflammatory cytokines (TNF-α, iNOS, and IL-6) and an M1 surface marker (CD86). The in vitro assays suggested that curcumin treatment suppressed the migration and invasion of the Cal27 cells induced by the M2-like macrophages. Mechanistically, the repolarization of TAMs may be attributed to the inhibition of monoamine oxidase A (MAO-A)/STAT6 signaling after curcumin treatment. Collectively, our results show that the anticancer effects of curcumin could be explained by reprogramming TAMs from a protumor phenotype towards an antitumor phenotype.
Collapse
Affiliation(s)
- Mingjing Jiang
- Liaoning Provincial Key Laboratory of Oral Diseases, Experimental Teaching Center, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Ying Qi
- Liaoning Provincial Key Laboratory of Oral Diseases, Experimental Teaching Center, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Wei Huang
- Liaoning Provincial Key Laboratory of Oral Diseases, Experimental Teaching Center, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Ying Lin
- Liaoning Provincial Key Laboratory of Oral Diseases, Experimental Teaching Center, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Bo Li
- Liaoning Provincial Key Laboratory of Oral Diseases, Experimental Teaching Center, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
- Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Department of Oral Anatomy and Physiology, Hospital of Stomatology, Jilin University, Changchun 130021, China
| |
Collapse
|
20
|
Gao J, Lu F, Yan J, Wang R, Xia Y, Wang L, Li L, Chang L, Li W. The role of radiotherapy-related autophagy genes in the prognosis and immune infiltration in lung adenocarcinoma. Front Immunol 2022; 13:992626. [PMID: 36311724 PMCID: PMC9606704 DOI: 10.3389/fimmu.2022.992626] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background There is a close relationship between radiotherapy and autophagy in tumors, but the prognostic role of radiotherapy-related autophagy genes (RRAGs) in lung adenocarcinoma (LUAD) remains unclear. Methods Data used in the current study were extracted from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Weighted gene co-expression network analysis (WGCNA) was executed to recognize module genes associated with radiotherapy. The differentially expressed genes (DEGs) between different radiotherapy response groups were filtered via edgeR package. The differentially expressed radiotherapy-related autophagy genes (DERRAGs) were obtained by overlapping the module genes, DEGs, and autophagy genes (ATGs). Then, prognostic autophagy genes were selected by Cox analyses, and a risk model and nomogram were subsequently built. Gene Set Enrichment Analysis (GSEA) and single-sample Gene Set Enrichment Analysis (ssGSEA) were performed to investigate potential mechanisms through which prognostic autophagy signatures regulate LUAD. Radiotherapy-resistant cell lines (A549IR and PC9IR) were established after exposure to hypo-fractionated irradiation. Ultimately, mRNA expression was validated by quantitative real-time PCR (qRT-PCR), and relative protein levels were measured in different cell lines by western blot. Results A total of 11 DERRAGs were identified in LUAD. After Cox analyses, SHC1, NAPSA, and AURKA were filtered as prognostic signatures in LUAD. Then, the risk score model was constructed using the prognostic signatures, which had a good performance in predicting the prognosis, as evidenced by receiver operating characteristics curves. Furthermore, Cox regression analyses demonstrated that risk score was deemed as an independent prognostic factor in LUAD. Moreover, GSEA and ssGSEA results revealed that prognostic RRAGs may regulate LUAD by modulating the immune microenvironment and affecting cell proliferation. The colony formation assay showed that the radiosensitivity of radiation-resistant cell lines was lower than that of primary cells. The western blot assay found that the levels of autophagy were elevated in the radiotherapy-resistant cell lines. Moreover, the expression of DERRAGs (SHC1, AURKA) was higher in the radiotherapy-resistant cells than in primary cells. Conclusion Our study explored the role of RRAGs in the prognosis of LUAD and identified three biomarkers. The findings enhanced the understanding of the relationship between radiotherapy, autophagy, and prognosis in LUAD and provided potential therapeutic targets for LUAD patients.
Collapse
Affiliation(s)
- Jingyan Gao
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, China
| | - Fei Lu
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, China
- Department of Oncology and Hematology, Southern Central Hospital of Yunnan Province, The First People’s Hospital of Honghe State, Mengzi, China
| | - Jiawen Yan
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, China
| | - Run Wang
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, China
| | - Yaoxiong Xia
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, China
| | - Li Wang
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, China
| | - Lan Li
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, China
| | - Li Chang
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, China
- *Correspondence: Wenhui Li, ; Li Chang,
| | - Wenhui Li
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, China
- *Correspondence: Wenhui Li, ; Li Chang,
| |
Collapse
|
21
|
Zhang Y, Feng Z, Liu J, Li H, Su Q, Zhang J, Huang P, Wang W, Liu J. Polarization of tumor-associated macrophages by TLR7/8 conjugated radiosensitive peptide hydrogel for overcoming tumor radioresistance. Bioact Mater 2022; 16:359-371. [PMID: 35386314 PMCID: PMC8965723 DOI: 10.1016/j.bioactmat.2021.12.033] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/29/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
Radioresistance reduces the antitumor efficiency of radiotherapy and further restricts its clinical application, which is mainly caused by the aggravation of immunosuppressive tumor microenvironment (ITM). Especially tumor-associated macrophages (TAMs) usually display the tumor-promoting M2 phenotype during high-dose fractional radiotherapy mediating radiotherapy resistance. Herein, the toll like receptor agonist TLR7/8a was conjugated with radiosensitive peptide hydrogel (Smac-TLR7/8 hydrogel) to regulate TAMs repolarization from M2 type into M1 type, thus modulating the ITM and overcoming the radioresistance. The Smac-TLR7/8 hydrogel was fabricated through self-assembly with nanofibrous morphology, porous structure and excellent biocompatibility. Upon γ-ray radiation, Smac-TLR7/8 hydrogel effectively polarized the macrophages into M1 type. Notably, combined with radiotherapy, TAMs repolarization regulated by Smac-TLR7/8 hydrogel could increase tumor necrosis factor secretion, activate antitumor immune response and effectively inhibit tumor growth. Moreover, TAMs repolarization rebuilt the ITM and elicited the immunogenic phenotypes in solid tumors, thus enhanced the PD1-blockade efficacy through increasing tumor infiltrating lymphocytes (TILs) and decreasing Treg cells in two different immune activity tumor mice models. Overall, this study substantiated that recruiting and repolarization of TAMs were critical in eliciting antitumor immune response and overcoming radioresistance, thus improving the efficacy of radiotherapy and immunotherapy.
Collapse
Affiliation(s)
- Yumin Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
| | - Zujian Feng
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300192, PR China
| | - Jinjian Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
| | - Hui Li
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
| | - Qi Su
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300192, PR China
| | - Jiamin Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300192, PR China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300192, PR China
- Corresponding author.
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
- Corresponding author.
| |
Collapse
|
22
|
Shi M, Chen Y, Ji D. The implications from the interplay of neoadjuvant chemoradiotherapy and the immune microenvironment in rectal cancer. Future Oncol 2022; 18:3229-3244. [PMID: 36017694 DOI: 10.2217/fon-2022-0061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neoadjuvant chemoradiotherapy (nCRT) is recommended for the treatment of locally advanced rectal cancer. Even though the combination of nCRT and immune checkpoint inhibitors (ICIs) has received much attention, the specific combination modes and dose fractions in radiotherapy (RT) are still indistinct. This review focuses on the immunological mechanism involved in nCRT, the clinical efficacy, the immunological effect of different combined strategies, concurrent or sequential nCRT plus ICIs, long-course RT and short-course RT. This review discusses the impact of nCRT on tumor immunity and summarizes the availability of different dose fractions in RT and distinct combined strategies, aiming at providing clues for optimal neoadjuvant therapy options that maximize efficacy and minimize side effects.
Collapse
Affiliation(s)
- Mengyuan Shi
- Key laboratory of Carcinogenesis & Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Haidian District, Beijing, 100142, China
| | - Yongkang Chen
- Key laboratory of Carcinogenesis & Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Haidian District, Beijing, 100142, China
| | - Dengbo Ji
- Key laboratory of Carcinogenesis & Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Haidian District, Beijing, 100142, China
| |
Collapse
|
23
|
Zhang Z, Liu X, Chen D, Yu J. Radiotherapy combined with immunotherapy: the dawn of cancer treatment. Signal Transduct Target Ther 2022; 7:258. [PMID: 35906199 PMCID: PMC9338328 DOI: 10.1038/s41392-022-01102-y] [Citation(s) in RCA: 270] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/19/2022] [Accepted: 06/30/2022] [Indexed: 11/09/2022] Open
Abstract
Radiotherapy (RT) is delivered for purposes of local control, but can also exert systemic effect on remote and non-irradiated tumor deposits, which is called abscopal effect. The view of RT as a simple local treatment has dramatically changed in recent years, and it is now widely accepted that RT can provoke a systemic immune response which gives a strong rationale for the combination of RT and immunotherapy (iRT). Nevertheless, several points remain to be addressed such as the interaction of RT and immune system, the identification of the best schedules for combination with immunotherapy (IO), the expansion of abscopal effect and the mechanism to amplify iRT. To answer these crucial questions, we roundly summarize underlying rationale showing the whole immune landscape in RT and clinical trials to attempt to identify the best schedules of iRT. In consideration of the rarity of abscopal effect, we propose that the occurrence of abscopal effect induced by radiation can be promoted to 100% in view of molecular and genetic level. Furthermore, the “radscopal effect” which refers to using low-dose radiation to reprogram the tumor microenvironment may amplify the occurrence of abscopal effect and overcome the resistance of iRT. Taken together, RT could be regarded as a trigger of systemic antitumor immune response, and with the help of IO can be used as a radical and systemic treatment and be added into current standard regimen of patients with metastatic cancer.
Collapse
Affiliation(s)
- Zengfu Zhang
- Department of Radiation Oncology, Shandong University Cancer Center, Yantai Road, No. 2999, Jinan, Shandong, China
| | - Xu Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road, No. 440, Jinan, Shandong, China
| | - Dawei Chen
- Department of Radiation Oncology, Shandong University Cancer Center, Yantai Road, No. 2999, Jinan, Shandong, China.
| | - Jinming Yu
- Department of Radiation Oncology, Shandong University Cancer Center, Yantai Road, No. 2999, Jinan, Shandong, China.
| |
Collapse
|
24
|
Yan D, Zhao Q, Du Z, Li H, Geng R, Yang W, Zhang X, Cao J, Yi N, Zhou J, Tang Z. Development and validation of an immune-related gene signature for predicting the radiosensitivity of lower-grade gliomas. Sci Rep 2022; 12:6698. [PMID: 35461367 PMCID: PMC9035187 DOI: 10.1038/s41598-022-10601-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/22/2022] [Indexed: 12/21/2022] Open
Abstract
Radiotherapy is an important treatment modality for lower-grade gliomas (LGGs) patients. This analysis was conducted to develop an immune-related radiosensitivity gene signature to predict the survival of LGGs patients who received radiotherapy. The clinical and RNA sequencing data of LGGs were obtained from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA). Lasso regression analyses were used to construct a 21-gene signature to identify the LGGs patients who could benefit from radiotherapy. Based on this radiosensitivity signature, patients were classified into a radiosensitive (RS) group and a radioresistant (RR) group. According to the Kaplan–Meier analysis results of the TCGA dataset and the two CGGA validation datasets, the RS group had a higher overall survival rate than that of the RR group. This gene signature was RT-specific and an independent prognostic indicator. The nomogram model performed well in predicting 3-, and 5-year survival of LGGs patients after radiotherapy by this gene signature and other clinical factors (age, sex, grade, IDH mutations, 1p/19q codeletion). In summary, this signature is a powerful supplement to the prognostic factors of LGGs patients with radiotherapy and may provide an opportunity to incorporate individual tumor biology into clinical decision making in radiation oncology.
Collapse
|
25
|
Hassel JC, Schank TE, Smetak H, Mühlbauer J, Salzmann M, Machiraju D, Menzer C, Lang K, König L, Haefner MF, Hülsmeyer I, Kohler C, Spang R, Enk A, Debus J, Beckhove P. Evaluation of radio-immunotherapy sequence on immunological responses and clinical outcomes in patients with melanoma brain metastases (ELEKTRA). Oncoimmunology 2022; 11:2066609. [PMID: 35481285 PMCID: PMC9037491 DOI: 10.1080/2162402x.2022.2066609] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In patients with melanoma brain metastases (MBM), a combination of radiotherapy (RT) with immune checkpoint inhibitors (ICI) is routinely used. However, the best sequence of radio-immunotherapy (RIT) remains unclear. In an exploratory phase 2 trial, MBM patients received RT (stereotactic or whole-brain radiotherapy depending on the number of MBM) combined with ipilimumab (ipi) ± nivolumab (nivo) in different sequencing (Rad-ICI or ICI-Rad). Comparators arms included patients treated with ipi-free systemic treatment or without RT (in MBM-free patients). The primary endpoints were radiological and immunological responses in the peripheral blood. Secondary endpoints were progression-free survival (PFS) and overall survival (OS). Of 106 screened, 92 patients were included in the study. Multivariate analysis revealed an advantage for patients starting with RT (Rad-ICI) for overall response rate (RR: p = .007; HR: 7.88 (95%CI: 1.76–35.27)) and disease control rate (DCR: p = .036; HR: 6.26 (95%CI: 1.13–34.71)) with a trend for a better PFS (p = .162; HR: 1.64 (95%CI: 0.8–3.3)). After RT plus two cycles of ipi-based ICI in both RIT sequences, increased frequencies of activated CD4, CD8 T cells and an increase in melanoma-specific T cell responses were observed in the peripheral blood. Lasso regression analysis revealed a significant clinical benefit for patients treated with Rad-ICI sequence and immunological features, including high frequencies of memory T cells and activated CD8 T cells in the blood. This study supports increasing evidence that sequencing RT followed by ICI treatment may have better effects on the immunological responses and clinical outcomes in MBM patients.
Collapse
Affiliation(s)
- Jessica C. Hassel
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg. Germany
| | - Timo E. Schank
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg. Germany
| | - Heiko Smetak
- Regensburg Center for Interventional Immunology, University Hospital Regensburg, Regensburg, Germany
| | - Jasmin Mühlbauer
- Regensburg Center for Interventional Immunology, University Hospital Regensburg, Regensburg, Germany
| | - Martin Salzmann
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg. Germany
| | - Devayani Machiraju
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg. Germany
| | - Christian Menzer
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg. Germany
| | - Kristin Lang
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Laila König
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Matthias F. Haefner
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Ingrid Hülsmeyer
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg. Germany
- The Immune Monitoring Unit, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Christian Kohler
- Statistical Bioinformatics Department, Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Rainer Spang
- Statistical Bioinformatics Department, Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Alexander Enk
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg. Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Philipp Beckhove
- Regensburg Center for Interventional Immunology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
26
|
Gao R, Shi GP, Wang J. Functional Diversities of Regulatory T Cells in the Context of Cancer Immunotherapy. Front Immunol 2022; 13:833667. [PMID: 35371055 PMCID: PMC8969660 DOI: 10.3389/fimmu.2022.833667] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/28/2022] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) are a subset of CD4+ T cells with their immunosuppressive activities to block abnormal or excessive immune responses to self and non-autoantigens. Tregs express the transcription factor Foxp3, maintain the immune homeostasis, and prevent the initiation of anti-tumor immune effects in various ways as their mechanisms to modulate tumor development. Recognition of different phenotypes and functions of intratumoral Tregs has offered the possibilities to develop therapeutic strategies by selectively targeting Tregs in cancers with the aim of alleviating their immunosuppressive activities from anti-tumor immune responses. Several Treg-based immunotherapeutic approaches have emerged to target cytotoxic T lymphocyte antigen-4, glucocorticoid-induced tumor necrosis factor receptor, CD25, indoleamine-2, 3-dioxygenase-1, and cytokines. These immunotherapies have yielded encouraging outcomes from preclinical studies and early-phase clinical trials. Further, dual therapy or combined therapy has been approved to be better choices than single immunotherapy, radiotherapy, or chemotherapy. In this short review article, we discuss our current understanding of the immunologic characteristics of Tregs, including Treg differentiation, development, therapeutic efficacy, and future potential of Treg-related therapies among the general cancer therapy.
Collapse
Affiliation(s)
- Ran Gao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Pan Y, Tang W, Fan W, Zhang J, Chen X. Development of nanotechnology-mediated precision radiotherapy for anti-metastasis and radioprotection. Chem Soc Rev 2022; 51:9759-9830. [DOI: 10.1039/d1cs01145f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Radiotherapy (RT), including external beam RT and internal radiation therapy, uses high-energy ionizing radiation to kill tumor cells.
Collapse
Affiliation(s)
- Yuanbo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Wei Tang
- Departments of Pharmacy and Diagnostic Radiology, Nanomedicine Translational Research Program, Faculty of Science and Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117544, Singapore
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
28
|
A Radioresponse-Related lncRNA Biomarker Signature for Risk Classification and Prognosis Prediction in Non-Small-Cell Lung Cancer. JOURNAL OF ONCOLOGY 2021; 2021:4338838. [PMID: 34594376 PMCID: PMC8478572 DOI: 10.1155/2021/4338838] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/06/2021] [Indexed: 12/11/2022]
Abstract
Purpose Radiotherapy resistance is now recognized as the major obstacle to the effective therapeutic management of non-small-cell lung cancer (NSCLC). As a single biomarker has limited effect in stratifying NSCLC patients, this research aimed to identify long non-coding RNAs (lncRNAs) correlated with radiotherapy response to ameliorate forecast of NSCLC prognosis. Methods In a cohort of NSCLC patients with radiotherapy history (n = 96) from TCGA, genetic data of lncRNA expression profiling were performed. To identify radioresponse-related lncRNA sets which dysregulated significantly between radiosensitive (RS) and radioresistant (RR) groups, differential expression analysis was carried out. Cox relative regression was implemented to set up a radioresponse-related risk model. Moreover, we adopted survival analysis to measure the predictive potentiality of the prognosis model. Results Four radioresponse-related lncRNAs (CASC19, LINC01977, LINC02471, and MAGI2-AS3) were screened to create a prognostic signature. Then, we described a lncRNA signature-based regulatory network and explored the correlation of the immune microenvironment and the signature. Additionally, in vitro assays uncovered inhibition of LINC01977 weakened radioresistance of NSCLC cells. Conclusion We provided a novel radioresponse-related lncRNAs signature with excellent clinical potency for an effective prognostic forecast of patients.
Collapse
|
29
|
Sia J, Hagekyriakou J, Chindris I, Albarakati H, Leong T, Schlenker R, Keam SP, Williams SG, Neeson PJ, Johnstone RW, Haynes NM. Regulatory T Cells Shape the Differential Impact of Radiation Dose-Fractionation Schedules on Host Innate and Adaptive Antitumor Immune Defenses. Int J Radiat Oncol Biol Phys 2021; 111:502-514. [PMID: 34023423 DOI: 10.1016/j.ijrobp.2021.05.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/04/2021] [Accepted: 05/13/2021] [Indexed: 12/29/2022]
Abstract
PURPOSE We examined how radiation dose per fraction (DPF) and total dose, as represented by biological effective dose (BED), can independently and differentially affect the immunomodulatory capacity of radiation therapy (RT). METHODS AND MATERIALS AT3-OVA mammary and MC38 colorectal tumors in C57BL/6 mice were irradiated with rationally selected dose-fractionation schedules, alone or with immune-modulating or -depleting agents. Tumor growth was monitored as a readout of therapeutic response. Flow cytometry and RNA sequencing of mouse tumors and analysis of transcriptomic data sets from irradiated human cancers were used to examine the immunomodulatory effects of the different radiation schedules. RESULTS In AT3-OVA tumors, radiation DPF rather than BED determined the ability of RT to evoke local antitumor CD8+ T cell responses and synergize with anti-PD-1 therapy. Natural killer cell-mediated control of irradiated tumors was more sensitive to radiation BED. Radiation-induced regulatory T cell (Treg) responses, which were detected in both mouse and human tumors, were a major factor underlying the differential activation of adaptive immunity by radiation DPF and the activity of natural killer cells during the early phase of response to RT. Targeted inhibition of Treg responses within irradiated tumors rescued and enhanced local tumor control by RT and permitted the generation of abscopal and immunologic memory responses, irrespective of radiation schedule. MC38 tumors did not support the induction of an amplified Treg response to RT and were highly vulnerable to its immunoadjuvant effects. CONCLUSIONS Local radiation-induced Treg responses are influenced by radiation schedule and tumor type and are a critical determinant of the immunoadjuvant potential of RT and its ability to synergize with T cell-targeted immunotherapy.
Collapse
Affiliation(s)
- Joseph Sia
- Translational Hematology Program, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia; Radiation Oncology Department, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Jim Hagekyriakou
- Physical Sciences Department, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Ioana Chindris
- Translational Hematology Program, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia
| | - Hassan Albarakati
- Physical Sciences Department, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Trevor Leong
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia; Radiation Oncology Department, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Ramona Schlenker
- Roche Pharmaceutical Research and Early Development, Roche Innovation Centre Munich, Penzberg, Germany
| | - Simon P Keam
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia; Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Australia; Cancer Immunology Research Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Scott G Williams
- Radiation Oncology Department, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Paul J Neeson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia; Cancer Immunology Research Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Ricky W Johnstone
- Translational Hematology Program, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia
| | - Nicole M Haynes
- Translational Hematology Program, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
30
|
Ukleja J, Kusaka E, Miyamoto DT. Immunotherapy Combined With Radiation Therapy for Genitourinary Malignancies. Front Oncol 2021; 11:663852. [PMID: 34041029 PMCID: PMC8141854 DOI: 10.3389/fonc.2021.663852] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy drugs have recently been approved by the Food and Drug Administration for the treatment of several genitourinary malignancies, including bladder cancer, renal cancer, and prostate cancer. Preclinical data and early clinical trial results suggest that immune checkpoint inhibitors can act synergistically with radiation therapy to enhance tumor cell killing at local irradiated sites and in some cases at distant sites through an abscopal effect. Because radiation therapy is commonly used in the treatment of genitourinary malignancies, there is great interest in testing the combination of immunotherapy with radiation therapy in these cancers to further improve treatment efficacy. In this review, we discuss the current evidence and biological rationale for combining immunotherapy with radiation therapy, as well as emerging data from ongoing and planned clinical trials testing the efficacy and tolerability of this combination in the treatment of genitourinary malignancies. We also outline outstanding questions regarding sequencing, dose fractionation, and biomarkers that remain to be addressed for the optimal delivery of this promising treatment approach.
Collapse
Affiliation(s)
- Jacob Ukleja
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Erika Kusaka
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - David T. Miyamoto
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Massachusetts General Hospital Cancer Center, Charlestown, MA, United States
| |
Collapse
|
31
|
Lumniczky K, Impens N, Armengol G, Candéias S, Georgakilas AG, Hornhardt S, Martin OA, Rödel F, Schaue D. Low dose ionizing radiation effects on the immune system. ENVIRONMENT INTERNATIONAL 2021; 149:106212. [PMID: 33293042 PMCID: PMC8784945 DOI: 10.1016/j.envint.2020.106212] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/20/2020] [Accepted: 09/03/2020] [Indexed: 05/03/2023]
Abstract
Ionizing radiation interacts with the immune system in many ways with a multiplicity that mirrors the complexity of the immune system itself: namely the need to maintain a delicate balance between different compartments, cells and soluble factors that work collectively to protect, maintain, and restore tissue function in the face of severe challenges including radiation damage. The cytotoxic effects of high dose radiation are less relevant after low dose exposure, where subtle quantitative and functional effects predominate that may go unnoticed until late after exposure or after a second challenge reveals or exacerbates the effects. For example, low doses may permanently alter immune fitness and therefore accelerate immune senescence and pave the way for a wide spectrum of possible pathophysiological events, including early-onset of age-related degenerative disorders and cancer. By contrast, the so called low dose radiation therapy displays beneficial, anti-inflammatory and pain relieving properties in chronic inflammatory and degenerative diseases. In this review, epidemiological, clinical and experimental data regarding the effects of low-dose radiation on the homeostasis and functional integrity of immune cells will be discussed, as will be the role of immune-mediated mechanisms in the systemic manifestation of localized exposures such as inflammatory reactions. The central conclusion is that ionizing radiation fundamentally and durably reshapes the immune system. Further, the importance of discovery of immunological pathways for modifying radiation resilience amongst other research directions in this field is implied.
Collapse
Affiliation(s)
- Katalin Lumniczky
- National Public Health Centre, Department of Radiation Medicine, Budapest, Albert Florian u. 2-6, 1097, Hungary.
| | - Nathalie Impens
- Belgian Nuclear Research Centre, Biosciences Expert Group, Boeretang 200, 2400 Mol, Belgium.
| | - Gemma Armengol
- Unit of Biological Anthropology, Department of Animal Biology, Plant Biology and Ecology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193-Bellaterra, Barcelona, Catalonia, Spain.
| | - Serge Candéias
- Université Grenoble-Alpes, CEA, CNRS, IRIG-LCBM, 38000 Grenoble, France.
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou 15780, Athens, Greece.
| | - Sabine Hornhardt
- Federal Office for Radiation Protection (BfS), Ingolstaedter Landstr.1, 85764 Oberschleissheim, Germany.
| | - Olga A Martin
- Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne 3052, Victoria, Australia.
| | - Franz Rödel
- Department of Radiotherapy and Oncology, University Hospital, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA.
| |
Collapse
|
32
|
Liu C, Piao J, Shang Z. Hyperprogressive disease after radiotherapy combined with anti-PD-1 therapy in renal cell carcinoma: a case report and review of the literature. BMC Urol 2021; 21:42. [PMID: 33743675 PMCID: PMC7981866 DOI: 10.1186/s12894-021-00813-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/16/2021] [Indexed: 01/10/2023] Open
Abstract
Background Studies have shown that immune checkpoint inhibitors (ICIs) have limited efficacy and can even increase tumour burden in short time periods. This is usually called hyperprogressive disease (HPD). To date, there are few reports regarding HPD; fewer have analysed the relationship between HPD and radiotherapy combined with ICIs, and their conclusions are controversial. Case presentation A 42-year-old woman was diagnosed with stage IV renal clear cell carcinoma. The patient had previously received sorafenib and pazopanib as first- and second-line therapies, respectively. She received radiotherapy combined with nivolumab. Eighteen days after administration of the third dose of nivolumab, the patient’s general condition deteriorated; this was associated with immune-related adverse events. Computed tomography showed that the diameter of left lung metastases had sharply increased. A biopsy of the lung metastasis showed no infiltration of lymphocytes. The patient’s general condition worsened and she died of the disease on the 70th day after administration of the third dose of nivolumab. Conclusions This report describes the development of HPD following the administration of radiotherapy combined with ICIs in a case of advanced renal cell carcinoma. The case indicates that radiotherapy may show bidirectional regulation effects on anti-tumour immune response. If the immunosuppressive function of radiotherapy is dominant, combined with ICIs, it could result in HPD. Supplementary Information The online version contains supplementary material available at 10.1186/s12894-021-00813-8.
Collapse
Affiliation(s)
- Chao Liu
- Department of Oncology, Zhuozhou Hospital, No.129, Fanyang Road, Zhuozhou City, Hebei Province, China
| | - Jingjing Piao
- College of Nursing, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
| | - Zhiyang Shang
- Department of Oncology, Zhuozhou Hospital, No.129, Fanyang Road, Zhuozhou City, Hebei Province, China.
| |
Collapse
|
33
|
Lei H, Shi M, Xu H, Bai S, Xiong X, Wei Q, Yang L. Combined Treatment of Radiotherapy and Immunotherapy for Urological Malignancies: Current Evidence and Clinical Considerations. Cancer Manag Res 2021; 13:1719-1731. [PMID: 33658847 PMCID: PMC7917304 DOI: 10.2147/cmar.s288337] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/23/2021] [Indexed: 02/05/2023] Open
Abstract
Although it has always been believed that radiation has immunosuppressive effects, more and more preclinical and clinical trials have shown that the combination of radiotherapy and immunotherapy has a potential synergistic effect to treat cancers including urological malignancies. When radiotherapy is combined with immunotherapy, improved prognosis has been observed in different urinary tumors. However, there is no standard treatment, such as the optimal dose/fractionation and the sequence of immunotherapy and radiotherapy. In this review, we discussed the effects of radiotherapy on the cancer immune system and emphasized the synergy of radiotherapy combined with immunotherapy. Although it has significantly improved the prognosis of tumors, there are still some unresolved questions about how to best use this combination in clinical practice. Ongoing trials will provide further information on the interaction of radiotherapy combined with immunotherapy, and are expected to guide clinical practice and improve clinical outcomes.
Collapse
Affiliation(s)
- Haoran Lei
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ming Shi
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hang Xu
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shengjiang Bai
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xingyu Xiong
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Lu Yang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
34
|
Kim N, Myoung Noh J, Lee W, Park B, Park H, Young Park J, Pyo H. Proton beam therapy reduces the risk of severe radiation-induced lymphopenia during chemoradiotherapy for locally advanced non-small cell lung cancer: A comparative analysis of proton versus photon therapy. Radiother Oncol 2020; 156:166-173. [PMID: 33359267 DOI: 10.1016/j.radonc.2020.12.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/23/2022]
Abstract
HYPOTHESIS We investigated differences in severe radiation-induced lymphopenia (SRL) after pencil beam scanning proton therapy (PBSPT) or intensity-modulated (photon) radiotherapy (IMRT) for patients with locally advanced non-small cell lung cancer. METHODS We retrospectively reviewed 223 patients who received definitive concurrent chemoradiotherapy with PBSPT (n = 29) or IMRT (n = 194). SRL was defined when ≥2 events of absolute lymphocyte counts (ALCs) of <200 cells/μL were observed in weekly laboratory tests conducted during treatment. Stepwise multivariate logistic regression with 10-fold cross-validation was performed to identify predictive values of SRL. Furthermore, 1:2 propensity score matching (PSM) analysis was performed between the PBSPT and IMRT groups. RESULTS Baseline ALC was comparable between the PBSPT and IMRT groups (median, 2130 vs. 2040 cells/μL; p = 0.983). Lung volumes receiving ≥ 5-20 GyE and the mean dose were significantly lower in patients receiving PBSPT than those receiving IMRT (p < 0.001). Among 72 (32.3%) patients with SRL; 69 (95.8%) and 3 (4.2%) patients were treated with IMRT and PBSPT, respectively. After multivariable analysis, PBSPT reduced SRL compared to IMRT (odds ratio [OR] 0.13, p = 0.003). Specifically, lung V5Gy were identified as the strongest predictor of SRL before (OR 1.11) and after PSM (OR, 1.07) (p < 0.05). With a median follow-up of 23.0 months, the 2-year overall survival in patients with SRL was worse than that those without SRL (63.4% vs. 79.9%; p = 0.003). CONCLUSIONS Reduced irradiated lung volumes of PBSPT consequently reduced SRL. In addition, lung V5Gy contributed to the SRL. Reduction of SRL through the optimized RT might be essential to improve the outcomes.
Collapse
Affiliation(s)
- Nalee Kim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jae Myoung Noh
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Woojin Lee
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Byoungsuk Park
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Heejoo Park
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ji Young Park
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hongryull Pyo
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
35
|
Yang Y, Ge H. Effective combinations of radiotherapy and immunotherapy in the treatment of esophageal squamous cell carcinoma. Future Oncol 2020; 16:2537-2549. [PMID: 33108227 DOI: 10.2217/fon-2020-0222] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The traditional treatments for esophageal squamous cell carcinoma include surgery and radiation as local therapies, then chemotherapy and targeted therapy as systemic treatments. These treatments, either alone or in combination, however, are not satisfactory because of limited efficacy and unfavorable toxicity, calling for new therapeutic strategies. In recent years, immunotherapy, a new weapon in the arsenal against cancer, has shown substantial clinical benefits in patients with esophageal squamous cell carcinoma, particularly ones with locally advanced or metastatic disease. Importantly, accumulating evidence suggests that traditional radiation therapy functions as a powerful adjuvant for immunotherapy by contributing to systemic antitumor immunity, resulting in reduced recurrence risk and improved survival of patients. Here the authors summarize the emerging data on immunotherapy- and radiation therapy-based treatment of esophageal squamous cell carcinoma and discuss the pros and cons of different combinations, aiming at a comprehensive understanding of the proper rationale for the design of effective therapeutic regimens.
Collapse
Affiliation(s)
- Yang Yang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, PR China
| | - Hong Ge
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, PR China
| |
Collapse
|
36
|
Krisnawan VE, Stanley JA, Schwarz JK, DeNardo DG. Tumor Microenvironment as a Regulator of Radiation Therapy: New Insights into Stromal-Mediated Radioresistance. Cancers (Basel) 2020; 12:cancers12102916. [PMID: 33050580 PMCID: PMC7600316 DOI: 10.3390/cancers12102916] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Cancer is multifaceted and consists of more than just a collection of mutated cells. These cancerous cells reside along with other non-mutated cells in an extracellular matrix which together make up the tumor microenvironment or tumor stroma. The composition of the tumor microenvironment plays an integral role in cancer initiation, progression, and response to treatments. In this review, we discuss how the tumor microenvironment regulates the response and resistance to radiation therapy and what targeted agents have been used to combat stromal-mediated radiation resistance. Abstract A tumor is a complex “organ” composed of malignant cancer cells harboring genetic aberrations surrounded by a stroma comprised of non-malignant cells and an extracellular matrix. Considerable evidence has demonstrated that components of the genetically “normal” tumor stroma contribute to tumor progression and resistance to a wide array of treatment modalities, including radiotherapy. Cancer-associated fibroblasts can promote radioresistance through their secreted factors, contact-mediated signaling, downstream pro-survival signaling pathways, immunomodulatory effects, and cancer stem cell-generating role. The extracellular matrix can govern radiation responsiveness by influencing oxygen availability and controlling the stability and bioavailability of growth factors and cytokines. Immune status regarding the presence of pro- and anti-tumor immune cells can regulate how tumors respond to radiation therapy. Furthermore, stromal cells including endothelial cells and adipocytes can modulate radiosensitivity through their roles in angiogenesis and vasculogenesis, and their secreted adipokines, respectively. Thus, to successfully eradicate cancers, it is important to consider how tumor stroma components interact with and regulate the response to radiation. Detailed knowledge of these interactions will help build a preclinical rationale to support the use of stromal-targeting agents in combination with radiotherapy to increase radiosensitivity.
Collapse
Affiliation(s)
- Varintra E. Krisnawan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jennifer A. Stanley
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; (J.A.S.); (J.K.S.)
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Julie K. Schwarz
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; (J.A.S.); (J.K.S.)
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David G. DeNardo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Correspondence:
| |
Collapse
|
37
|
Steenbrugge J, De Jaeghere EA, Meyer E, Denys H, De Wever O. Splenic Hematopoietic and Stromal Cells in Cancer Progression. Cancer Res 2020; 81:27-34. [PMID: 32998999 DOI: 10.1158/0008-5472.can-20-2339] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/31/2020] [Accepted: 09/24/2020] [Indexed: 11/16/2022]
Abstract
Tumor-derived secretory factors orchestrate splenic hematopoietic and stromal cells to fuel metastasis. The spleen acts as a reservoir site for hematopoietic stem and progenitor cells, which are rapidly exploited as myeloid-derived suppressor cells at the cost of tumor-reactive lymphoid cells. Splenic erythroid progenitor cells and mesenchymal stromal cells contribute directly and indirectly to both tumor immune escape and the metastatic cascade. Animal models provide valuable mechanistic insights, but their translation to a clinical setting highlights specific challenges and open issues. In this review, we envision the exploitation of the spleen as a source for novel biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
- Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Emiel A De Jaeghere
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Medical Oncology, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Gynecologic Pelvic Oncology Network Ghent (GYPON), Ghent, Belgium
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Hannelore Denys
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Medical Oncology, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Gynecologic Pelvic Oncology Network Ghent (GYPON), Ghent, Belgium
| | - Olivier De Wever
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| |
Collapse
|
38
|
Daro-Faye M, Kassouf W, Souhami L, Marcq G, Cury F, Niazi T, Sargos P. Combined radiotherapy and immunotherapy in urothelial bladder cancer: harnessing the full potential of the anti-tumor immune response. World J Urol 2020; 39:1331-1343. [PMID: 32915313 PMCID: PMC7484608 DOI: 10.1007/s00345-020-03440-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/03/2020] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Radiotherapy (RT), as part of trimodal therapy, is an attractive alternative treatment in patients with urothelial muscle-invasive bladder cancer (MIBC). There is accumulating evidence suggesting the immunomodulatory effects of RT and its potential synergy when combined with immunotherapy. The aim of this review was to report on the most recent advances on this combination, including the mechanisms of RT immunomodulation, practical approach to combining RT and immunotherapy, and ongoing clinical trials in bladder cancer. METHODS Using the PubMed database, we identified articles published between March 2004 and April 2020 on the combination of RT with immunotherapy in localized or metastatic MIBC. A search of the Clinicaltrials.gov and Clinicaltrialsregister.eu/ retrieved ongoing clinical trials on the topic as well. RESULTS Combination of RT with immunotherapy leads to immunogenic cell death and an increase in immune markers thus leading to improved tumor control. For localized MIBC, there are safety concerns related to the use of concurrent immunotherapy with hypofractionated RT, thus neoadjuvant or adjuvant immunotherapy is preferred. In the metastatic setting, the combination of multi-site RT with SBRT-like doses (≥ 6 Gy per fraction) and concurrent immunotherapy seems most efficacious at harnessing the abscopal effect. At least 25 clinical trials combining immunotherapy and RT in MIBC are currently ongoing and will answer pending questions on safety, efficacy, and practical considerations on RT scheduling, fractionation, and targets volumes. CONCLUSION RT has the potential to synergize with immunotherapy to improve oncological outcomes in patient with localized or metastatic MIBC. Clinical trials results are eagerly awaited.
Collapse
Affiliation(s)
- Mame Daro-Faye
- Department of Radiation Oncology, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Wassim Kassouf
- Department of Urology, McGill University Health Center, Montreal, QC, Canada
| | - Luis Souhami
- Fellow of the American Society for Radiation Oncology (FASTRO), Department of Radiation Oncology, McGill University Health Centre, Montreal, QC, Canada
| | - Gautier Marcq
- Department of Urology, McGill University Health Center, Montreal, QC, Canada.,Univ. Lille, Inserm, CHU Lille, U1189-ONCO-THAI-Laser Assisted Therapies and Immunotherapies for Oncology, 59000, Lille, France.,Department of Urology, Claude Huriez Hospital, CHU Lille, 59000, Lille, France
| | - Fabio Cury
- Fellow of the American Society for Radiation Oncology (FASTRO), Department of Radiation Oncology, McGill University Health Centre, Montreal, QC, Canada
| | - Tamim Niazi
- Department of Radiation Oncology, Jewish General Hospital, Montreal, QC, Canada
| | - Paul Sargos
- Department of Radiation Oncology, Jewish General Hospital, Montreal, QC, Canada. .,Department of Radiation Oncology, Comprehensive Cancer Center, Institut Bergonie, 33076, Bordeaux Cedex, France.
| |
Collapse
|
39
|
Guo T, Zou L, Ni J, Zhou Y, Ye L, Yang X, Zhu Z. Regulatory T Cells: An Emerging Player in Radiation-Induced Lung Injury. Front Immunol 2020; 11:1769. [PMID: 32849634 PMCID: PMC7417370 DOI: 10.3389/fimmu.2020.01769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/01/2020] [Indexed: 12/25/2022] Open
Abstract
Regulatory T cells (Tregs), which have long been recognized as essential regulators of both inflammation and autoimmunity, also impede effective antitumor immune response due to their immunosuppressive properties. Combined radiotherapy and immunotherapeutic interventions focusing on the removal of Tregs have recently garnered interest as a promising strategy to reverse immunosuppression. Meanwhile, Tregs are emerging as a key player in the pathogenesis of radiation-induced lung injury (RILI), a frequent and potentially life-threatening complication of thoracic radiotherapy. Recognition of the critical role of Tregs in RILI raises the important question of whether radiotherapy combined with Treg-targeting immunotherapy offers any beneficial effects in the protection of normal lung tissue. This present review focuses on the contributions of Tregs to RILI, with particular emphasis on the suspected differential role of Tregs in the pneumonitic phase and fibrotic phase of RILI. We also introduce recent progress on the potential mechanisms by which Tregs modulate RILI and the crosstalk among Tregs, other infiltrating T cells, fibrocytes, and resident epithelial cells driving disease pathogenesis. Finally, we discuss whether Tregs also hold promise as a potential target for immunotherapeutic interventions for RILI.
Collapse
Affiliation(s)
- Tiantian Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liqing Zou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yue Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Luxi Ye
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
40
|
Pellizzon ACA. Lattice radiation therapy - its concept and impact in the immunomodulation cancer treatment era. ACTA ACUST UNITED AC 2020; 66:728-731. [PMID: 32696876 DOI: 10.1590/1806-9282.66.6.728] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/08/2019] [Indexed: 04/05/2023]
Abstract
Voluminous tumors represent a challenge in radiation oncology, particularly when surgical resection is not possible. Lattice radiotherapy (LTR) is a technique that may provide equivalent or superior clinical response in the management of large tumors while limiting toxicity to adjacent normal tissues. LRT can precisely deliver inhomogeneous high doses of radiation to different areas within the gross tumor volumes (GTV). The dosimetric characteristic of LTR is defined by the ratio of the valley dose (lower doses - cold spots) and the peak doses, also called vertex (higher doses - hot spots), or the valley-to-peak dose ratio. The valley-to-peak ratio thereby quantifies the degree of spatial fractionation. LRT delivers high doses of radiation without exceeding the tolerance of adjacent critical structures. Radiobiological experiments support the role of radiation-induced bystander effects, vascular alterations, and immunologic interactions in areas subject to low dose radiation. The technological advancements continue to expand in Radiation Oncology, bringing new safety opportunities of treatment for bulky lesions.
Collapse
|
41
|
Piper M, Mueller AC, Karam SD. The interplay between cancer associated fibroblasts and immune cells in the context of radiation therapy. Mol Carcinog 2020; 59:754-765. [PMID: 32363633 DOI: 10.1002/mc.23205] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023]
Abstract
Fibroblasts are a key component of the tumor microenvironment (TME) that can serve as a scaffold for tumor cell migration and augment the tumor's ability to withstand harsh conditions. When activated by external or endogenous stimuli, normal fibroblasts become cancer associated fibroblasts (CAFs), a heterogeneous group of stromal cells in the tumor that are phenotypically and epigenetically different from normal fibroblasts. Dynamic crosstalk between cancer cells, immune cells, and CAFs through chemokines and surface signaling makes the TME conducive to tumor growth. When activated, CAFs promote tumorigenesis and metastasis through several phenomena including regulation of tumor immunity, metabolic reprogramming of the TME, extracellular matrix remodeling and contraction, and induction of therapeutic resistance. Ionizing radiation (radiation theraphy [RT]) is a potent immunological stimulant that has been shown to increase cytotoxic Teff infiltration and IFN-I stimulated genes. RT, however, is unable to overcome the infiltration and activation of immunosuppressive cells which can contribute to tumor progression. Another paradox of RT is that, while very effective at killing cancer cells, it can contribute to the formation of CAFs. This review examines how the interplay between CAFs and immune cells during RT contributes to organ fibrosis, immunosuppression, and tumor growth. We focus on targeting mechanistic pathways of CAF formation as a potentially effective strategy not only for preventing organ fibrosis, but also in hampering tumor progression in response to RT.
Collapse
Affiliation(s)
- Miles Piper
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Adam C Mueller
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
42
|
Chen VE, Greenberger BA, Taylor JM, Edelman MJ, Lu B. The Underappreciated Role of the Humoral Immune System and B Cells in Tumorigenesis and Cancer Therapeutics: A Review. Int J Radiat Oncol Biol Phys 2020; 108:38-45. [PMID: 32251756 DOI: 10.1016/j.ijrobp.2020.03.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 03/21/2020] [Indexed: 02/07/2023]
Abstract
The advent of immunotherapy has ushered in a new era in both cancer research and cancer treatment strategies. Published reviews have described potential mechanisms for therapeutic synergisms from the combination of radiation therapy and immunotherapy, largely overlooking the role of humoral immunity by only focusing on cellular immunity. Given that these 2 branches of the immune system are highly interdependent, in this review we detail both what has already been established regarding the role of humoral immunity in cancer and propose potential avenues that are ripe for further investigation and potential clinical applications.
Collapse
Affiliation(s)
- Victor E Chen
- Department of Radiation Oncology, Sidney Kimmel Medical College & Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Benjamin A Greenberger
- Department of Radiation Oncology, Sidney Kimmel Medical College & Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - James M Taylor
- Department of Radiation Oncology, Sidney Kimmel Medical College & Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Martin J Edelman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Bo Lu
- Department of Radiation Oncology, Sidney Kimmel Medical College & Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
43
|
Lambin P, Lieverse RIY, Eckert F, Marcus D, Oberije C, van der Wiel AMA, Guha C, Dubois LJ, Deasy JO. Lymphocyte-Sparing Radiotherapy: The Rationale for Protecting Lymphocyte-rich Organs When Combining Radiotherapy With Immunotherapy. Semin Radiat Oncol 2020; 30:187-193. [PMID: 32381298 PMCID: PMC8412054 DOI: 10.1016/j.semradonc.2019.12.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is now strong clinical and preclinical evidence that lymphocytes, for example, CD8+ T cells, are key effectors of immunotherapy and that irradiation of large blood vessels, the heart, and lymphoid organs (including nodes, spleen, bones containing bone marrow, and thymus in children) causes transient or persistent lymphopenia. Furthermore, there is extensive clinical evidence, across multiple cancer sites and treatment modalities, that lymphopenia correlates strongly with decreased overall survival. At the moment, we lack quantitative evidence to establish the relationship between dose-volume and dose-rate to critical normal structures and lymphopenia. Therefore, we propose that data should be systematically recorded to characterise a possible quantitative relationship. This might enable us to improve the efficacy of radiotherapy and develop strategies to predict and prevent treatment-related lymphopenia. In anticipation of more quantitative data, we recommend the application of the principle of As Low As Reasonably Achievable to lymphocyte-rich regions for radiotherapy treatment planning to reduce the radiation doses to these structures, thus moving toward "Lymphocyte-Sparing Radiotherapy."
Collapse
Affiliation(s)
- Philippe Lambin
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.
| | - Relinde I Y Lieverse
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Franziska Eckert
- Department of Radiation Oncology, University Hospital and Medical Faculty Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Damiënne Marcus
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Cary Oberije
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Alexander M A van der Wiel
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY
| | - Ludwig J Dubois
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
44
|
Huang J, Zhang D, Bai Y, Yang P, Xing L, Yu J. A 2AR Antagonism with DZD2269 Augments Antitumor Efficacy of Irradiation in Murine Model. J Cancer 2020; 11:3685-3692. [PMID: 32284765 PMCID: PMC7150468 DOI: 10.7150/jca.43966] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 02/27/2020] [Indexed: 11/13/2022] Open
Abstract
Accumulated extracellular adenosine suppresses antitumor immunity via adenosine 2A receptor (A2AR). Blockade of A2AR with DZD2269 can inhibit phosphorylation of cAMP response element-binding protein mediated by adenosine analogue in vitro and in vivo. Irradiation can cause the release of adenosine and lead to a rapid increase in free extracellular adenosine in the tumour area. DZD2269, a novel A2AR Antagonism, induces incomplete antitumor responses in multiple syngeneic mouse tumour models. Combining DZD2269 with IR can induce a synergistic anticancer effect. IR increases the infiltration of various subtypes of T cells, including CD4+, CD8+ and Foxp3+ T cells, into the tumour area. Combining IR and DZD2269 improves the tumour immune microenvironment, leading to suppressed infiltration of regulatory T (Treg) cells and enhanced IFN-γ expression by tumour-infiltrating lymphocytes. The results support the use of A2AR antagonism with DZD2269 as a therapeutic strategy for monotherapy or combination therapy with IR.
Collapse
Affiliation(s)
- Jiaqi Huang
- Department of Clinical Medicine, Shandong University, Jinan, Shandong 250000, P.R. China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Di Zhang
- Department of Clinical Medicine, Shandong University, Jinan, Shandong 250000, P.R. China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Yu Bai
- Dizal (Jiangsu) Pharmaceutical Co., Ltd., Wuxi, Jiangsu 214028, P.R. China
| | - Pamela Yang
- Dizal (Jiangsu) Pharmaceutical Co., Ltd., Wuxi, Jiangsu 214028, P.R. China
| | - Ligang Xing
- Department of Clinical Medicine, Shandong University, Jinan, Shandong 250000, P.R. China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Jinming Yu
- Department of Clinical Medicine, Shandong University, Jinan, Shandong 250000, P.R. China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
45
|
Lai X, Friedman A. Mathematical modeling of cancer treatment with radiation and PD-L1 inhibitor. SCIENCE CHINA MATHEMATICS 2020; 63:465-484. [DOI: 10.1007/s11425-019-1648-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/27/2020] [Indexed: 01/04/2025]
|
46
|
Colbert LE, Jhingran A. Immunotherapy and Radiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1244:205-213. [DOI: 10.1007/978-3-030-41008-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
47
|
Verma A, Mathur R, Farooque A, Kaul V, Gupta S, Dwarakanath BS. T-Regulatory Cells In Tumor Progression And Therapy. Cancer Manag Res 2019; 11:10731-10747. [PMID: 31920383 PMCID: PMC6935360 DOI: 10.2147/cmar.s228887] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/06/2019] [Indexed: 12/24/2022] Open
Abstract
Regulatory T cells (Tregs) are important members of the immune system regulating the host responses to infection and neoplasms. Tregs prevent autoimmune disorders by protecting the host-cells from an immune response, related to the peripheral tolerance. However, tumor cells use Tregs as a shield to protect themselves against anti-tumor immune response. Thus, Tregs are a hurdle in achieving the complete potential of anti-cancer therapies including immunotherapy. This has prompted the development of novel adjuvant therapies that obviate their negative effects thereby enhancing the therapeutic efficacy. Our earlier studies have shown the efficacy of the glycolytic inhibitor, 2-deoxy-D-glucose (2-DG) by reducing the induced Tregs pool and enhance immune stimulation as well as local tumor control. These findings have suggested its potential for enhancing the efficacy of immunotherapy, besides radiotherapy and chemotherapy. This review provides a brief account of the current status of Tregs as a component of the immune-biology of tumors and various preclinical and clinical strategies pursued to obviate the limitations imposed by them in achieving therapeutic efficacy.
Collapse
Affiliation(s)
- Amit Verma
- Armed Forces Radiobiology Research Institute, Uniformed Services University, Bethesda, MD, USA
| | - Rohit Mathur
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Vandana Kaul
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Seema Gupta
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | | |
Collapse
|
48
|
Targets for improving tumor response to radiotherapy. Int Immunopharmacol 2019; 76:105847. [PMID: 31466051 DOI: 10.1016/j.intimp.2019.105847] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/18/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023]
|
49
|
Liao XY, Liu CY, He JF, Wang LS, Zhang T. Combination of checkpoint inhibitors with radiotherapy in esophageal squamous cell carcinoma treatment: A novel strategy. Oncol Lett 2019; 18:5011-5021. [PMID: 31612012 PMCID: PMC6781725 DOI: 10.3892/ol.2019.10893] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023] Open
Abstract
Despite the rapid development of numerous types of treatment, including radiotherapy (RT) as the main strategy, esophageal squamous cell carcinoma (ESCC) has a poor prognosis. Recent studies demonstrated that immunotherapy can improve the survival of patients with locally advanced and metastatic ESCC. Furthermore, previous studies reported that the expression of programmed death-ligand 1 is significantly associated with esophageal cancer prognosis. At present, several ongoing clinical trials have extended the use of immunotherapy from palliative and salvage treatments to neoadjuvant treatment with concurrent chemoradiation. The first- or second-line treatments were used to explore antitumor efficacy with reduced adverse events. The combination of RT and immunotherapy can exert a local therapeutic effect and improve the function of the immune system, enhancing antitumor efficacy. This review investigated the role of immunotherapy and radiotherapy in ESCC and described the potential efficacy of combining immunotherapy with radiotherapy in ESCC.
Collapse
Affiliation(s)
- Xiu-Yong Liao
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
- Department of Oncology, Chongqing Qianjiang Central Hospital, Chongqing 409000, P.R. China
| | - Chao-Yuan Liu
- Department of Neurosurgery, Chongqing Qianjiang Central Hospital, Chongqing 409000, P.R. China
| | - Jian-Feng He
- Department of General Surgery, Chongqing Qianjiang Central Hospital, Chongqing 409000, P.R. China
| | - Li-Shu Wang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Tao Zhang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
50
|
Caetano MS, Younes AI, Barsoumian HB, Quigley M, Menon H, Gao C, Spires T, Reilly TP, Cadena AP, Cushman TR, Schoenhals JE, Li A, Nguyen QN, Cortez MA, Welsh JW. Triple Therapy with MerTK and PD1 Inhibition Plus Radiotherapy Promotes Abscopal Antitumor Immune Responses. Clin Cancer Res 2019; 25:7576-7584. [PMID: 31540976 DOI: 10.1158/1078-0432.ccr-19-0795] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/10/2019] [Accepted: 09/17/2019] [Indexed: 01/07/2023]
Abstract
PURPOSE Radiotherapy (RT) traditionally has been used for local tumor control in the treatment of cancer. The recent discovery that radiotherapy can have anticancer effects on the immune system has led to recognition of its ability to sensitize the tumor microenvironment to immunotherapy. However, radiation can also prompt adverse immunosuppressive effects that block aspects of systemic response at other tumor sites. Our hypothesis was that inhibition of the MER proto-oncogene tyrosine kinase (MerTK) in combination with anti-programmed cell death-1 (α-PD1) checkpoint blockade will enhance immune-mediated responses to radiotherapy. EXPERIMENTAL DESIGN We tested the efficacy of this triple therapy (Radiation + α-PD1 + α-MerTK mAbs) in 129Sv/Ev mice with bilateral lung adenocarcinoma xenografts. Primary tumors were treated with stereotactic radiotherapy (36 Gy in 3 12-Gy fractions), and tumors were monitored for response. RESULTS The triple therapy significantly delayed abscopal tumor growth, improved survival rates, and reduced numbers of lung metastases. We further found that the triple therapy increased the activated CD8+ and NK cells populations measured by granzyme B expression with upregulation of CD8+CD103+ tissue-resident memory cells (TRM) within the abscopal tumor microenvironment relative to radiation only. CONCLUSIONS The addition of α-PD1 + α-MerTK mAbs to radiotherapy could alter the cell death to be more immunogenic and generate adaptive immune response via increasing the retention of TRM cells in the tumor islets of the abscopal tumors which was proven to play a major role in survival of non-small cell lung cancer patients.
Collapse
Affiliation(s)
- Mauricio S Caetano
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ahmed I Younes
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Michael Quigley
- Bristol-Myers Squibb (BMS), Redwood City, California and Princeton, New Jersey
| | - Hari Menon
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chan Gao
- Bristol-Myers Squibb (BMS), Redwood City, California and Princeton, New Jersey
| | - Thomas Spires
- Bristol-Myers Squibb (BMS), Redwood City, California and Princeton, New Jersey
| | - Timothy P Reilly
- Bristol-Myers Squibb (BMS), Redwood City, California and Princeton, New Jersey
| | - Alexandra P Cadena
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Taylor R Cushman
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.,University of Arizona College of Medicine-Phoenix, Phoenix, Arizona
| | - Jonathan E Schoenhals
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.,University of Texas Southwestern Medical School, Dallas, Texas
| | - Ailin Li
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Radiation Oncology, First Hospital of China Medical University, China
| | - Quynh-Nhu Nguyen
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria Angelica Cortez
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James W Welsh
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|