1
|
Shi X, Zhao H, Yu J, Cai P, Zhou S, Yang N, Li D. Changes in PD-1 expression on T lymphocyte subsets and related immune indicators before and after definitive chemoradiotherapy for esophageal squamous cell carcinoma. Ann Med 2025; 57:2445190. [PMID: 39713872 DOI: 10.1080/07853890.2024.2445190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/29/2024] [Accepted: 11/30/2024] [Indexed: 12/24/2024] Open
Abstract
OBJECTIVE This study aimed to observe the dynamic changes in the expression of T lymphocytes, natural killer (NK) cells, and PD-1 in patients with first-diagnosed esophageal squamous cell carcinoma (ESCC) before and after chemoradiotherapy (CRT) and evaluate the impact of PD-1 expression in peripheral blood on the short-term outcome of patients with ESCC. PATIENTS AND METHODS Seventy-three patients with ESCC who were treated with definitive CRT were enrolled. Before and after CRT, flow cytometry was used to detect thePD-1 expression in the peripheral blood and related immune indicators. Peripheral blood from 10 healthy individuals was used as control. RESULTS The percentages of CD3+ (p = 0.018), CD4+ (p < 0.001), and CD8+ T cells (p < 0.001); NK cells (p = 0.009); and the CD4+/CD8+ ratio (p < 0.001), as well as PD-1+CD3+ (p < 0.001), PD-1+CD4+ (p < 0.001), and PD-1+CD8+ (p < 0.001) T cells, before CRT significantly differed from those in the post-CRT group. The percentages of PD-1+CD8+ T cells differed significantly between the radiotherapy alone and CRT groups (p < 0.05). PD-1 expression in CD3+, CD4+, and CD8+ T cells significantly decreased in patients achieving overall response rate (all p < 0.05). Compared with those in the incomplete response group, PD-1+CD8+ T cells significantly decreased in the CR group (p < 0.05). CONCLUSION CRT aggravated immunosuppression and increased PD-1 expression in T lymphocyte subsets in patients with ESCC, possibly related to the radiation field. PD-1 expression in T lymphocyte subsets can predict short-term outcomes in patients and provide a theoretical basis for the sequential application of PD-1 immunosuppressants after radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Xueling Shi
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Hongyu Zhao
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Jiaqi Yu
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Peng Cai
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Shixiang Zhou
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Ning Yang
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Duojie Li
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
2
|
Wei T, Lei M, Jiang H, Cai J, Peng Q, Wei Y, Chen Z, Geng J, Ren F, Chen C, Yang Z, Zhang Y, Chu Z, Jia H, Yin Z, Zhao T. Attenuated Salmonella carrying IL-21 overexpression plasmid enhances radiotherapy efficacy in a preclinical model of melanoma. Int Immunopharmacol 2025; 154:114590. [PMID: 40174337 DOI: 10.1016/j.intimp.2025.114590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
Melanoma, known for its aggressive behavior and tendency to metastasize to the brain and lungs, is a formidable challenge in oncology. Radiotherapy is a potent treatment for localized solid tumors, effective against both intracranial and extracranial metastases. Yet, some melanoma patients exhibit substantial resistance to radiotherapy, with the underlying mechanisms of this resistance remaining elusive. While radiotherapy can stimulate the infiltration of immune cells, thereby triggering a range of immunostimulatory effects, it can also suppress the tumor microenvironment (TME), limiting its effectiveness. In physiological conditions, cytokines inhibit the activity of immunosuppressive cells through paracrine and autocrine signaling, while also activating immune cells to boost antitumor responses. Here, we found that Interleukin (IL)-21 expression was higher in the mice with good radiotherapy response to melanoma than in the mice with poor radiotherapy response. Interestingly, we also observed the higher infiltration of M2 TAMs and lower CD8+ T cells in the group with poor radiotherapy response. To tackle this issue, we explored the therapeutic potential of a plasmid encoding IL-21, delivered via attenuated Salmonella, in mice bearing melanomas. Our findings revealed that IL-21 administration significantly reduced M2 TAMs infiltration and enhanced CD8+ T cells infiltration and granzyme B (GZMB) expression within melanoma tumors. Most importantly, the combination of IL-21 with radiotherapy led to markedly tumor reduction compared to either treatment alone. This research highlights the potential of IL-21 as a valuable adjunct to radiotherapy in the treatment of melanoma, presenting a promising strategy for enhancing antitumor immune responses and optimizing patient outcomes.
Collapse
Affiliation(s)
- Tian Wei
- Department of Immunology, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Henan International Joint Laboratory of Immunity and Targeted Therapy for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, PR China
| | - Mengyu Lei
- Department of Immunology, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China
| | - Hanyu Jiang
- Department of Immunology, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China
| | - Jingjing Cai
- Department of Immunology, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China
| | - Qi Peng
- Department of Immunology, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China
| | - Yuqing Wei
- Department of Immunology, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China
| | - Zhihan Chen
- Department of Immunology, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China
| | - Jiaxin Geng
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China
| | - Feng Ren
- Henan International Joint Laboratory of Immunity and Targeted Therapy for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, PR China
| | - Caili Chen
- Department of Immunology, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China
| | - Zishan Yang
- Department of Immunology, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, PR China
| | - Zhili Chu
- Henan International Joint Laboratory of Immunity and Targeted Therapy for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, PR China
| | - Huijie Jia
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China.
| | - Zhinan Yin
- Department of Immunology, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Henan International Joint Laboratory of Immunity and Targeted Therapy for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, PR China.
| | - Tiesuo Zhao
- Department of Immunology, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang 453000, Henan, PR China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang 453000, Henan, PR China.
| |
Collapse
|
3
|
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
|
4
|
Muhammad FA, Adhab AH, Mahdi MS, Jain V, Ganesan S, Bhanot D, Naidu KS, Kaur S, Mansoor AS, Radi UK, Abd NS, Kariem M. Unveiling Novel Targets in Lung Tumors for Enhanced Radiotherapy Efficacy: A Comprehensive Review. J Biochem Mol Toxicol 2025; 39:e70180. [PMID: 39987513 DOI: 10.1002/jbt.70180] [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: 10/23/2024] [Revised: 12/06/2024] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
Radiotherapy is a cornerstone of lung cancer management, though its efficacy is frequently undermined by intrinsic and acquired radioresistance. This review examines the complexity of lung tumors, highlighting their potential as a reservoir of novel targets for radiosensitization. Ionizing radiation (IR) primarily exerts its effects through oxidative damage and DNA double-strand breaks (DSBs). Lung cancer cells, however, develop mutations that enhance DNA damage response (DDR) and suppress cell death pathways. Additionally, interactions between tumor cells and tumor microenvironment (TME) components-including immune cells, stromal cells, and molecular mediators such as cytokines, chemokines, and growth factors-contribute to resistance against IR. Understanding these intricate relationships reveals potential targets to improve radiotherapy outcomes. Promising targets include DDR pathways, immunosuppressive cells and molecules, hypoxia, proangiogenic mediators, and other key signaling pathways. This review discusses emerging strategies, such as combining radiotherapy with immunomodulators, hypoxia and proangiogenic inhibitors, DDR-targeting agents, and other innovative approaches. By offering a comprehensive analysis of the lung TME, this review underscores opportunities to enhance radiotherapy effectiveness through targeted radiosensitization strategies.
Collapse
Affiliation(s)
| | | | | | - Vicky Jain
- Department of Chemistry, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, India
| | - Deepak Bhanot
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, India
| | - Sharnjeet Kaur
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, India
| | | | - Usama Kadem Radi
- Collage of Pharmacy, National University of Science and Technology, Nasiriyah, Iraq
| | - Nasr Saadoun Abd
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
5
|
Rafiq Z, Kang M, Barsoumian HB, Manzar GS, Hu Y, Leuschner C, Huang A, Masrorpour F, Lu W, Puebla-Osorio N, Welsh JW. Enhancing immunotherapy efficacy with synergistic low-dose radiation in metastatic melanoma: current insights and prospects. J Exp Clin Cancer Res 2025; 44:31. [PMID: 39881333 PMCID: PMC11781074 DOI: 10.1186/s13046-025-03281-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
Recent advances in oncology research have highlighted the promising synergy between low-dose radiation therapy (LDRT) and immunotherapies, with growing evidence highlighting the unique benefits of the combination. LDRT has emerged as a potent tool for stimulating the immune system, triggering systemic antitumor effects by remodeling the tumor microenvironment. Notably, LDRT demonstrates remarkable efficacy even in challenging metastatic sites such as the liver (uveal) and brain (cutaneous), particularly in advanced melanoma stages. The increasing interest in utilizing LDRT for secondary metastatic sites of uveal, mucosal, or cutaneous melanomas underscores its potential efficacy in combination with various immunotherapies. This comprehensive review traverses the journey from laboratory research to clinical applications, elucidating LDRT's immunomodulatory role on the tumor immune microenvironment (TIME) and systemic immune responses. We meticulously examine the preclinical evidence and ongoing clinical trials, throwing light on the promising prospects of LDRT as a complementary therapy in melanoma treatment. Furthermore, we explore the challenges associated with LDRT's integration into combination therapies, addressing crucial factors such as optimal dosage, fractionation, treatment frequency, and synergy with other pharmacological agents. Considering its low toxicity profile, LDRT presents a compelling case for application across multiple lesions, augmenting the antitumor immune response in poly-metastatic disease scenarios. The convergence of LDRT with other disciplines holds immense potential for developing novel radiotherapy-combined modalities, paving the way for more effective and personalized treatment strategies in melanoma and beyond. Moreover, the dose-related toxicities of immunotherapies may be reduced by synergistic amplification of antitumor efficacy with LDRT.
Collapse
Affiliation(s)
- Zahid Rafiq
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79968, USA
| | - Mingyo Kang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gohar S Manzar
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yun Hu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Carola Leuschner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ailing Huang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Fatemeh Masrorpour
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Weiqin Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79968, USA
| | - Nahum Puebla-Osorio
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
6
|
Cao Y, Zhao X, Miao Y, Wang X, Deng D. How the Versatile Self-Assembly in Drug Delivery System to Afford Multimodal Cancer Therapy? Adv Healthc Mater 2025; 14:e2403715. [PMID: 39587000 DOI: 10.1002/adhm.202403715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/04/2024] [Indexed: 11/27/2024]
Abstract
The rapid development of self-assembly technology during the past few decades has effectively addressed plenty of the issues associated with carrier-based drug delivery systems, such as low loading efficiency, complex fabrication processes, and inherent toxicity of carriers. The integration of nanoscale delivery systems with self-assembly techniques has enabled efficient and targeted self-administration of drugs, enhanced bioavailability, prolonged circulation time, and controllable drug release. Concurrently, the limitations of single-mode cancer treatment, including low bioavailability, poor therapeutic outcomes, and significant side effects, have highlighted the urgent need for multimodal combined antitumor therapies. Set against the backdrop of multimodal cancer therapy, this review summarizes the research progress and applications of a large number of self-assembled drug delivery platforms, including natural small molecule self-assembled, carrier-free self-assembled, amphiphilic polymer-based self-assembled, peptide-based self-assembled, and metal-based self-assembled nano drug delivery systems. This review particularly analyzes the latest advances in the application of self-assembled nano drug delivery platforms in combined antitumor therapies mediated by chemotherapy, phototherapy, radiotherapy, sonodynamic therapy, and immunotherapy, providing innovative research insights for further optimization and expansion of self-assembled nano drug delivery systems in the clinical translation and development of antitumor combined therapy.
Collapse
Affiliation(s)
- Yuqi Cao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaomin Zhao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuhang Miao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xin Wang
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Dawei Deng
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
7
|
Wang G, Man Y, Cao K, Zhao L, Lun L, Chen Y, Zhao X, Wang X, Zhang L, Hao C. An immune-related gene pair signature predicts the prognosis and immunotherapeutic response in glioblastoma. Heliyon 2024; 10:e39025. [PMID: 39435104 PMCID: PMC11492119 DOI: 10.1016/j.heliyon.2024.e39025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024] Open
Abstract
Background Glioblastoma (GBM) has the feature of aggressive growth and high rates of recurrence. Immunotherapy was not included in standard therapy for GBM due to lacking the predictive biomarkers. In the present study, we performed an immune-related gene pair (IRGP) signature to predict the prognosis and immunotherapy response of GBM. Methods A total of 160 GBM patients from TCGA were included. ssGSEA was conducted to evaluate the immune infiltration level. Univariate Cox, LASSO regression analysis, ROC analysis, and Kaplan-Meier survival analysis were applied to construct and evaluate the risk model. Moreover, the association between immune infiltration and the risk score was assessed. Finally, the expression of immune checkpoints between different risk groups was explored. Results According to the normal/tumor, high-/low-immunity group, we identified 125 differentially expressed immune-related genes. Subsequently, a prognostic model including 22 IRGPs was established. The area under the ROC curve to predict 1, 3, and 5-year was 0.811, 0.958, and 0.99 respectively. According to the optimal cut-off value of the 3-year ROC curve, patients were classified into high- and low-risk groups. The Kaplan-Meier analysis result indicated that patients in the low-risk group have longer survival time. The risk score was an independent prognostic predictor (P < 0.001). Moreover, PDCD1 was positively correlated with the risk score (P < 0.01). We also found that patients with high PDCD1 expression had worse survival. Conclusions The IRGP signature was built to predict the prognosis of GBM patients. This signature can serve as a tool to predict the response to immunotherapy in GBM.
Collapse
Affiliation(s)
- Gang Wang
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Department of Radiation Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingchun Man
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin, China
| | - Kui Cao
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lihong Zhao
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lixin Lun
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yiyang Chen
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xinyu Zhao
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xueying Wang
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lijie Zhang
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin, China
| | - Chuncheng Hao
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
8
|
Tang Q, Li L, Wang R, Jin X, Jia X, Zhu Y, Sun X, Zhong J, Xie H, Da Y, Zu L, Xu S. Constructing a folate metabolism gene signature for predicting prognosis in pulmonary neuroendocrine carcinomas. J Cancer 2024; 15:6256-6272. [PMID: 39513106 PMCID: PMC11540503 DOI: 10.7150/jca.102186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/30/2024] [Indexed: 11/15/2024] Open
Abstract
Folate metabolism is a crucial biological process in cell proliferation and exhibits its pro-tumorigenic functions in multiple tumor types. However, its role in pulmonary neuroendocrine carcinomas remains uncertain. Folate metabolism related genes were obtained from previous studies, and the gene expression data and clinical data were collected from GEO database. The expression patterns of folate metabolism related genes were measured across normal and tumor tissues. We subsequently assessed their prognostic role using Kaplan-Meier and univariate Cox regression analysis. The core genes were isolated from 16 prognostic genes through four algorithms. Based on the expression of core genes, patients were divided into two clusters employing consensus clustering algorithm. Furthermore, we evaluated immune infltration level, biological mechanisms, and drug sensitivity. ALDH1L2 was finally identified through qRT-PCR and its pro-tumorigenic function was confirmed via in vitro experiments. The expression patterns of 26 folate metabolism related genes were evaluated between normal lung tissues and PNEC tumor tissues, and 20 of them exhibited differential expression. All of folate metabolism related genes were related to the prognosis of PNECS and 16 genes were identified as prognostic genes. Using SVM-RFE, RF, Xgboost and LASSO algorithm, three core genes were isolated from 16 prognostic genes. Based on the expression patterns of core genes, PNECs patients were divided into two clusters through consensus clustering algorithm. Cluster 1 was characterized by the worse survival, higher immune infiltration level, and sensitivity to chemotherapy. Compared with the HBEC cells, ALDH1L2 was notably overexpressed in NCI-H446 cells (SCLC cell line). ALDH1L2 knockdown significantly repressed the proliferation and migration capacity of tumor cells and increased the cell proportion in S phase. Our results indicated that folate metabolism gene signature is a reliable biomarker for PNECs. Classification based on this signature could be utilized to guide the treatment of PNECs patients and improve its prognosis.
Collapse
Affiliation(s)
- Quanying Tang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Luoyi Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Ruiyao Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Xin Jin
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xuewang Jia
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yifan Zhu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiaoyue Sun
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jianguo Zhong
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Huangsheng Xie
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yurong Da
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Lingling Zu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Song Xu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
9
|
An R, Wu C, Tang C, Zhang C, Han F, Xu Z, Zou Y, Wang J, Yuan Z, Jiang S, Liu L, Huang C, Tao Z. Blockade of CD73 potentiates radiotherapy antitumor immunity and abscopal effects via STING pathway. Cell Death Discov 2024; 10:404. [PMID: 39285178 PMCID: PMC11405876 DOI: 10.1038/s41420-024-02171-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Abstract
Radiotherapy (RT) is a crucial treatment for colorectal cancer (CRC) patients, but it often fails to induce systemic antitumor immunity. CD73, an immunomodulatory factor, is upregulated after RT and associated with poor prognosis in CRC patients. This study aims to elucidate the mechanisms driving RT-induced CD73 upregulation in CRC and investigate how combining RT with CD73 blockade stimulates immune responses and induces abscopal effects. Findings revealed that RT-induced CD73 upregulation is mediated by the ataxia telangiectasia and Rad3-related (ATR) pathway and correlated with RT tolerance, as demonstrated through flow cytometry, immunofluorescence, and Western Blotting. Using flow cytometry and multicolor immunofluorescence, experiments demonstrated that in CRC subcutaneous tumor models, combination therapy reduces the infiltration of myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), and regulatory T cells (Tregs) while increasing dendritic cells (DCs) and CD8 + T cells, resulting in superior antitumor responses. Additionally, results from flow cytometry, Western Blot, and RNA sequencing demonstrated that combination therapy enhances the antigen-presenting ability of DCs and activates tumor antigen-specific CD8 + T cells, improving their function and delaying their depletion. The activation of the cGAS-STING and IFN-I pathways is crucial for this effect. In summary, the integration of RT with CD73 blockade effectively reverses the immunosuppressive TME and invigorates CD8 + T cell-driven, specific antitumor immune responses. These insights shed fresh light on the mechanisms governing the synergistic modulation of immunity by RT and CD73 blockade in CRC, offering promising avenues for the advancement of therapeutic strategies against CRC.
Collapse
Affiliation(s)
- Ran An
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin and Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Chao Wu
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin and Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Cunyu Tang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin and Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Chen Zhang
- Department of Anesthesiology, The First Central Clinical School, Tianjin Medical University, Tianjin, PR China
| | - Feiru Han
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin and Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Zeen Xu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin and Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Yiping Zou
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin and Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Jun Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin and Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Zhiyong Yuan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin and Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Shengpeng Jiang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin and Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Lijie Liu
- Department of Oncology, First Hospital of Qinhuangdao, Qinhuangdao, PR China
| | - Chongbiao Huang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin and Tianjin's Clinical Research Center for Cancer, Tianjin, PR China.
| | - Zhen Tao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin and Tianjin's Clinical Research Center for Cancer, Tianjin, PR China.
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
10
|
Wang X, Zhang H, XinZhang, Liu Y. Abscopal effect: from a rare phenomenon to a new frontier in cancer therapy. Biomark Res 2024; 12:98. [PMID: 39228005 PMCID: PMC11373306 DOI: 10.1186/s40364-024-00628-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Radiotherapy (RT) controls local lesions, meantime it has the capability to induce systemic response to inhibit distant, metastatic, non-radiated tumors, which is referred to as the "abscopal effect". It is widely recognized that radiotherapy can stimulate systemic immune response. This provides a compelling theoretical basis for the combination of immune therapy combined with radiotherapy(iRT). Indeed, this phenomenon has also been observed in clinical treatment, bringing significant clinical benefits to patients, and a series of basic studies are underway to amplify this effect. However, the molecular mechanisms of immune response induced by RT, determination of the optimal treatment regimen for iRT, and how to amplify the abscopal effect. In order to amplify and utilize this effect in clinical management, these key issues require to be well addressed; In this review, we comprehensively summarize the growing consensus and emphasize the emerging limitations of enhancing the abscopal effect with radiotherapy or immunotherapy. Finally, we discuss the prospects and barriers to the current clinical translational applications.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Haoyu Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - XinZhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| |
Collapse
|
11
|
Jiao X, Hong H, Cai W. Nanoscale Radiotheranostics for Cancer Treatment: From Bench to Bedside. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2006. [PMID: 39407431 PMCID: PMC11486289 DOI: 10.1002/wnan.2006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 08/31/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Abstract
In recent years, the application of radionuclides-containing nanomaterials in cancer treatment has garnered widespread attention. The diversity of nanomaterials allows researchers to selectively combine them with appropriate radionuclides for biomedical purposes, addressing challenges faced by peptides, small molecules, or antibodies used for radionuclide labeling. However, with advantages come challenges, and nanoradionuclides still encounter significant issues during clinical translation. This review summarized the recent progress of nanosized radionuclides for cancer treatment or diagnosis. The discussion began with representative radionuclides and the methods of incorporating them into nanomaterial structures. Subsequently, new combinations of nanomaterials and radionuclides, along with their applications, were introduced to demonstrate their future trends. The benefits of nanoradionuclides included optimized pharmacokinetic properties, enhanced disease-targeting efficacy, and synergistic application with other treatment techniques. Besides, the basic rule of this section was to summarize how these nanoradionuclides can truly impact the diagnosis and therapy of various cancer types. In the last part, the focus was devoted to the nanoradionuclides currently applicable in clinics and how to address the existing issues and problems based on our knowledge.
Collapse
Affiliation(s)
- Xiaodan Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Hao Hong
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, USA
| |
Collapse
|
12
|
Kluge A, Baum RP, Bitterlich N, Kulkarni HR, Schorr-Neufing U, van Echteld CJA. Immune Response to Molecular Radiotherapy with 177Lu-DOTATOC: Predictive Value of Blood Cell Counts for Therapy Outcome. Cancer Biother Radiopharm 2024; 39:541-550. [PMID: 38905126 DOI: 10.1089/cbr.2024.0031] [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: 06/23/2024] Open
Abstract
Purpose: In a prior, retrospective study, 76% of patients with advanced neuroendocrine tumors undergoing 177Lu-DOTATOC molecular radiotherapy (MRT) showed their best response within 8 months from the first MRT cycle. In 24% of patients, latency was much greater up to >22 months after the first cycle, and long after near-complete decay of 177Lu from the last cycle. An immune response induced by MRT seems a likely explanation. As a crude measure of immunocompetence, the authors investigated whether blood cell counts (BCCs) may have predictive value for MRT outcome with 177Lu-DOTATOC. Methods: 56 Patients with neuroendocrine tumors (NET) were administered 177Lu-DOTATOC (mean 2.1 cycles; range 1-4) with median radioactivity of 7.0 GBq/cycle at 3-month intervals. Patients' BCCs were evaluated for four responder categories: CR, PR, SD, and PD (RECIST 1.1). Furthermore, baseline BCCs were correlated with progression-free survival (PFS). Finally, BCCs of patients with (PMT+) and without prior medical therapy (PMT-) were compared. Results: Significant differences between responder categories were found for baseline hemoglobin (Hb), erythrocytes, neutrophils, lymphocytes, neutrophil/lymphocyte ratio (NLR), platelet/lymphocyte ratio (PLR), and LEHN-score, integrating lymphocyte, erythrocyte, and neutrophil counts, and Hb level, but not for leukocytes and platelets. LEHN-score yielded an almost complete separation between CR and PD groups. In analogy, PFS times showed significant correlations with baseline Hb, erythrocytes, neutrophils, lymphocytes, NLR, PLR, and LEHN-score, the LEHN-score showing the strongest correlation, but not with leukocytes and platelets. For PMT- patients, median PFS was 34.5 months, compared with 20.8 months in PMT+ patients, with corresponding baseline lymphocyte (32.1 ± 9.6% vs. 24.5 ± 11.6%, p = 0.028) and neutrophil (54.9 ± 11.6% vs. 63.5 ± 13.7%, p = 0.039) counts. Conclusion: These findings emphasize the significance of an immune response to MRT for obtaining optimal therapy efficacy and support concepts to enhance the immune response of less immunocompetent patients before MRT. It seems advisable to avoid prior or concomitant immunosuppressant medical therapy.
Collapse
Affiliation(s)
- Andreas Kluge
- ABX-CRO Advanced Pharmaceutical Services, Dresden, Germany
| | - Richard P Baum
- Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka, Bad Berka, Germany
- CURANOSTICUM Wiesbaden-Frankfurt-Advanced Theranostics Center for Radiomolecular Precision Oncology, HELIOS DKD Klinik, Wiesbaden, Germany
| | | | - Harshad R Kulkarni
- Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka, Bad Berka, Germany
- BAMF Health, Grand Rapids, Michigan, USA
| | | | - Cees J A van Echteld
- ABX-CRO Advanced Pharmaceutical Services, Dresden, Germany
- Helacor Consultancy, Hillegom, The Netherlands
| |
Collapse
|
13
|
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
|
14
|
Qiu L, Yang Z, Jia G, Liang Y, Du S, Zhang J, Liu M, Zhao X, Jiao S. Clinical significance and immune landscape of a novel immune cell infiltration-based prognostic model in lung adenocarcinoma. Heliyon 2024; 10:e33109. [PMID: 38988583 PMCID: PMC11234107 DOI: 10.1016/j.heliyon.2024.e33109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/08/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024] Open
Abstract
Tumor-infiltrating immune cells (TICs) play a central role in the tumor microenvironment, which can reflect the host anti-tumor immune response. However, few studies have explored TICs in predicting the prognosis of lung adenocarcinoma (LUAD). In our study, we enrolled 2470 LUAD patients from TCGA and GEO databases, and the normalized enrichment scores for 65 immune cell types were quantified for each patient. An immune-related risk score (IRRS) was built on the basis of 17 selected TICs using LASSO regression analysis, and the results showed that high-risk patients were correlated with shorter survival time for the LUAD cohorts. Correlation analyses between IRRS and clinical characteristics were also evaluated to validate the clinical use of IRRS. In addition, we analyzed the differences in the distribution of immune cell infiltration and immunoregulatory gene expression, which may facilitate individual immunotherapy. Based on the above result, we conclude that IRRS can act as a powerful predictor for risk stratification and prognosis prediction, and may facilitate the decision-making process for LUAD patients.
Collapse
Affiliation(s)
- Lupeng Qiu
- Department of Medical Oncology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Graduate Administration, Chinese PLA General Hospital, Beijing, China
| | - Zizhong Yang
- School of Medicine, Nankai University, Tianjin, China
| | - Guhe Jia
- School of Medicine, Nankai University, Tianjin, China
| | - Yanjie Liang
- Department of Medical Oncology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Sicheng Du
- Department of Medical Oncology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Graduate Administration, Chinese PLA General Hospital, Beijing, China
| | - Jian Zhang
- Department of Medical Oncology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Minglu Liu
- Department of Medical Oncology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiao Zhao
- Department of Medical Oncology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shunchang Jiao
- Department of Medical Oncology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Graduate Administration, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
15
|
Xu YY, Bai RX, Zhang QR, Zhang S, Zhang JH, Du SY. A comprehensive analysis of GAS2 family members identifies that GAS2L1 is a novel biomarker and promotes the proliferation of hepatocellular carcinoma. Discov Oncol 2024; 15:220. [PMID: 38858234 PMCID: PMC11164853 DOI: 10.1007/s12672-024-01083-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a common primary liver cancer with a high incidence and mortality. Members of the growth-arresting-specific 2 (GAS2) family are involved in various biological processes in human malignancies. To date, there is only a limited amount of information available about the expression profile and clinical importance of GAS2 family in HCC. In this study, we found that GAS2L1 and GAS2L3 were distinctly upregulated in HCC specimens compared to non-tumor specimens. Pan-cancer assays indicated that GAS2L1 and GAS2L3 were highly expressed in most cancers. The Pearson's correlation revealed that the expressions of GAS2, GAS2L1 and GAS2L2 were negatively associated with methylation levels. Survival assays indicated that GAS2L1 and GAS2L3 were independent prognostic factors for HCC patients. Immune cell infiltration analysis revealed that GAS2, GAS2L1 and GAS2L3 were associated with several immune cells. Finally, we confirmed that GAS2L1 was highly expressed in HCC cells and its knockdown suppressed the proliferation of HCC cells. Taken together, our findings suggested the expression patterns and prognostic values of GAS2 members in HCC, providing insights for further study of the GAS2 family as sensitive diagnostic and prognostic markers for HCC.
Collapse
Affiliation(s)
- Ying-Ying Xu
- Department of Gastroenterology, China-Japan Friendship Hospital, No. 2, Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Ru-Xue Bai
- Department of Gastroenterology, China-Japan Friendship Hospital, No. 2, Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Qing-Rui Zhang
- Department of Gastroenterology, China-Japan Friendship Hospital, No. 2, Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Shuang Zhang
- Department of Gastroenterology, China-Japan Friendship Hospital, No. 2, Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Jun-Hai Zhang
- Department of Gastroenterology, China-Japan Friendship Hospital, No. 2, Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
- Graduate School, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, People's Republic of China
| | - Shi-Yu Du
- Department of Gastroenterology, China-Japan Friendship Hospital, No. 2, Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China.
| |
Collapse
|
16
|
Fan J, Zhu J, Zhu H, Xu H. Potential therapeutic targets in myeloid cell therapy for overcoming chemoresistance and immune suppression in gastrointestinal tumors. Crit Rev Oncol Hematol 2024; 198:104362. [PMID: 38614267 DOI: 10.1016/j.critrevonc.2024.104362] [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: 11/18/2023] [Revised: 03/26/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024] Open
Abstract
In the tumor microenvironment (TME), myeloid cells play a pivotal role. Myeloid-derived immunosuppressive cells, including tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), are central components in shaping the immunosuppressive milieu of the tumor. Within the TME, a majority of TAMs assume an M2 phenotype, characterized by their pro-tumoral activity. These cells promote tumor cell growth, angiogenesis, invasion, and migration. In contrast, M1 macrophages, under appropriate activation conditions, exhibit cytotoxic capabilities against cancer cells. However, an excessive M1 response may lead to pro-tumoral inflammation. As a result, myeloid cells have emerged as crucial targets in cancer therapy. This review concentrates on gastrointestinal tumors, detailing methods for targeting macrophages to enhance tumor radiotherapy and immunotherapy sensitivity. We specifically delve into monocytes and tumor-associated macrophages' various functions, establishing an immunosuppressive microenvironment, promoting tumorigenic inflammation, and fostering neovascularization and stromal remodeling. Additionally, we examine combination therapeutic strategies.
Collapse
Affiliation(s)
- Jiawei Fan
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - Jianshu Zhu
- Department of Spine Surgery, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - He Zhu
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - Hong Xu
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China.
| |
Collapse
|
17
|
Shi Z, Hu C, Zheng X, Sun C, Li Q. Feedback loop between hypoxia and energy metabolic reprogramming aggravates the radioresistance of cancer cells. Exp Hematol Oncol 2024; 13:55. [PMID: 38778409 PMCID: PMC11110349 DOI: 10.1186/s40164-024-00519-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Radiotherapy is one of the mainstream approaches for cancer treatment, although the clinical outcomes are limited due to the radioresistance of tumor cells. Hypoxia and metabolic reprogramming are the hallmarks of tumor initiation and progression and are closely linked to radioresistance. Inside a tumor, the rate of angiogenesis lags behind cell proliferation, and the underdevelopment and abnormal functions of blood vessels in some loci result in oxygen deficiency in cancer cells, i.e., hypoxia. This prevents radiation from effectively eliminating the hypoxic cancer cells. Cancer cells switch to glycolysis as the main source of energy, a phenomenon known as the Warburg effect, to sustain their rapid proliferation rates. Therefore, pathways involved in metabolic reprogramming and hypoxia-induced radioresistance are promising intervention targets for cancer treatment. In this review, we discussed the mechanisms and pathways underlying radioresistance due to hypoxia and metabolic reprogramming in detail, including DNA repair, role of cancer stem cells, oxidative stress relief, autophagy regulation, angiogenesis and immune escape. In addition, we proposed the existence of a feedback loop between energy metabolic reprogramming and hypoxia, which is associated with the development and exacerbation of radioresistance in tumors. Simultaneous blockade of this feedback loop and other tumor-specific targets can be an effective approach to overcome radioresistance of cancer cells. This comprehensive overview provides new insights into the mechanisms underlying tumor radiosensitivity and progression.
Collapse
Affiliation(s)
- Zheng Shi
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Cuilan Hu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaogang Zheng
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chao Sun
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
18
|
Zhu L, Yu X, Tang X, Hu C, Wu L, Liu Y, Zhou Q. Evolving landscape of treatments targeting the microenvironment of liver metastases in non-small cell lung cancer. Chin Med J (Engl) 2024; 137:1019-1032. [PMID: 38251678 PMCID: PMC11062672 DOI: 10.1097/cm9.0000000000002981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Indexed: 01/23/2024] Open
Abstract
ABSTRACT Liver metastases (LMs) are common in lung cancer. Despite substantial advances in diagnosis and treatment, the survival rate of patients with LM remains low as the immune-suppressive microenvironment of the liver allows tumor cells to evade the immune system. The impact of LMs on the outcomes of immune checkpoint inhibitors in patients with solid tumors has been the main focus of recent translational and clinical research. Growing evidence indicates that the hepatic microenvironment delivers paracrine and autocrine signals from non-parenchymal and parenchymal cells. Overall, these microenvironments create pre- and post-metastatic conditions for the progression of LMs. Herein, we reviewed the epidemiology, physiology, pathology and immunology, of LMs associated with non-small cell lung cancer and the role and potential targets of the liver microenvironment in LM in each phase of metastasis. Additionally, we reviewed the current treatment strategies and challenges that should be overcome in preclinical and clinical investigations. These approaches target liver elements as the basis for future clinical trials, including combinatorial interventions reported to resolve hepatic immune suppression, such as immunotherapy plus chemotherapy, immunotherapy plus radiotherapy, immunotherapy plus anti-angiogenesis therapy, and surgical resection.
Collapse
Affiliation(s)
- Lingling Zhu
- Lung Cancer Center, Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xianzhe Yu
- Department of Gastrointestinal Surgery, Chengdu Second People’s Hospital, Chengdu, Sichuan 610041, China
| | - Xiaojun Tang
- Lung Cancer Center, Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chenggong Hu
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lei Wu
- Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yanyang Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qinghua Zhou
- Lung Cancer Center, Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
19
|
Peng Y, Liang S, Meng QF, Liu D, Ma K, Zhou M, Yun K, Rao L, Wang Z. Engineered Bio-Based Hydrogels for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313188. [PMID: 38362813 DOI: 10.1002/adma.202313188] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/01/2024] [Indexed: 02/17/2024]
Abstract
Immunotherapy represents a revolutionary paradigm in cancer management, showcasing its potential to impede tumor metastasis and recurrence. Nonetheless, challenges including limited therapeutic efficacy and severe immune-related side effects are frequently encountered, especially in solid tumors. Hydrogels, a class of versatile materials featuring well-hydrated structures widely used in biomedicine, offer a promising platform for encapsulating and releasing small molecule drugs, biomacromolecules, and cells in a controlled manner. Immunomodulatory hydrogels present a unique capability for augmenting immune activation and mitigating systemic toxicity through encapsulation of multiple components and localized administration. Notably, hydrogels based on biopolymers have gained significant interest owing to their biocompatibility, environmental friendliness, and ease of production. This review delves into the recent advances in bio-based hydrogels in cancer immunotherapy and synergistic combinatorial approaches, highlighting their diverse applications. It is anticipated that this review will guide the rational design of hydrogels in the field of cancer immunotherapy, fostering clinical translation and ultimately benefiting patients.
Collapse
Affiliation(s)
- Yuxuan Peng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Qian-Fang Meng
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Kongshuo Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Mengli Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Kaiqing Yun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
20
|
Qiu L, Ji H, Wang K, Liu W, Huang Q, Pan X, Ye H, Li Z, Chen G, Xing X, Dong X, Tang R, Xu H, Liu J, Cai Z, Liu X. TLR3 activation enhances abscopal effect of radiotherapy in HCC by promoting tumor ferroptosis. EMBO Mol Med 2024; 16:1193-1219. [PMID: 38671318 PMCID: PMC11098818 DOI: 10.1038/s44321-024-00068-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Radiotherapy (RT) has been reported to induce abscopal effect in advanced hepatocellular carcinoma (HCC), but such phenomenon was only observed in sporadic cases. Here, we demonstrated that subcutaneous administration of Toll-like receptor 3 (TLR3) agonist poly(I:C) could strengthen the abscopal effect during RT through activating tumor cell ferroptosis signals in bilateral HCC subcutaneous tumor mouse models, which could be significantly abolished by TLR3 knock-out or ferroptosis inhibitor ferrostatin-1. Moreover, poly(I:C) could promote the presentation of tumor neoantigens by dendritic cells to enhance the recruitment of activated CD8+ T cells into distant tumor tissues for inducing tumor cell ferroptosis during RT treatment. Finally, the safety and feasibility of combining poly(I:C) with RT for treating advanced HCC patients were further verified in a prospective clinical trial. Thus, enhancing TLR3 signaling activation during RT could provide a novel strategy for strengthening abscopal effect to improve the clinical benefits of advanced HCC patients.
Collapse
Affiliation(s)
- Liman Qiu
- College of Chemical Engineering, Fuzhou University, Fuzhou, P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, P. R. China
| | - Hongbing Ji
- Radiotherapy Department, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
| | - Kai Wang
- Radiotherapy Department, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
| | - Wenhan Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
| | - Qizhen Huang
- Radiotherapy Department, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
| | - Xinting Pan
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, P. R. China
| | - Honghao Ye
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
| | - Zhenli Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, P. R. China
| | - Geng Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, P. R. China
| | - Xiaohua Xing
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, P. R. China
| | - Xiuqing Dong
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, P. R. China
| | - Ruijing Tang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, P. R. China
| | - Haipo Xu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, P. R. China
| | - Jingfeng Liu
- College of Chemical Engineering, Fuzhou University, Fuzhou, P. R. China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China.
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, P. R. China.
- Mengchao Med-X Center, Fuzhou University, Fuzhou, P. R. China.
| | - Xiaolong Liu
- College of Chemical Engineering, Fuzhou University, Fuzhou, P. R. China.
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China.
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, P. R. China.
- Mengchao Med-X Center, Fuzhou University, Fuzhou, P. R. China.
| |
Collapse
|
21
|
Noh SU, Lim J, Shin SW, Kim Y, Park WY, Batinic-Haberle I, Choi C, Park W. Single-Cell Profiling Reveals Immune-Based Mechanisms Underlying Tumor Radiosensitization by a Novel Mn Porphyrin Clinical Candidate, MnTnBuOE-2-PyP 5+ (BMX-001). Antioxidants (Basel) 2024; 13:477. [PMID: 38671924 PMCID: PMC11047573 DOI: 10.3390/antiox13040477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Manganese porphyrins reportedly exhibit synergic effects when combined with irradiation. However, an in-depth understanding of intratumoral heterogeneity and immune pathways, as affected by Mn porphyrins, remains limited. Here, we explored the mechanisms underlying immunomodulation of a clinical candidate, MnTnBuOE-2-PyP5+ (BMX-001, MnBuOE), using single-cell analysis in a murine carcinoma model. Mice bearing 4T1 tumors were divided into four groups: control, MnBuOE, radiotherapy (RT), and combined MnBuOE and radiotherapy (MnBuOE/RT). In epithelial cells, the epithelial-mesenchymal transition, TNF-α signaling via NF-кB, angiogenesis, and hypoxia-related genes were significantly downregulated in the MnBuOE/RT group compared with the RT group. All subtypes of cancer-associated fibroblasts (CAFs) were clearly reduced in MnBuOE and MnBuOE/RT. Inhibitory receptor-ligand interactions, in which epithelial cells and CAFs interacted with CD8+ T cells, were significantly lower in the MnBuOE/RT group than in the RT group. Trajectory analysis showed that dendritic cells maturation-associated markers were increased in MnBuOE/RT. M1 macrophages were significantly increased in the MnBuOE/RT group compared with the RT group, whereas myeloid-derived suppressor cells were decreased. CellChat analysis showed that the number of cell-cell communications was the lowest in the MnBuOE/RT group. Our study is the first to provide evidence for the combined radiotherapy with a novel Mn porphyrin clinical candidate, BMX-001, from the perspective of each cell type within the tumor microenvironment.
Collapse
Affiliation(s)
- Sun Up Noh
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Republic of Korea; (S.U.N.); (S.-W.S.); (Y.K.)
- Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Jinyeong Lim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea; (J.L.); (W.-Y.P.)
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Sung-Won Shin
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Republic of Korea; (S.U.N.); (S.-W.S.); (Y.K.)
| | - Yeeun Kim
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Republic of Korea; (S.U.N.); (S.-W.S.); (Y.K.)
| | - Woong-Yang Park
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea; (J.L.); (W.-Y.P.)
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA;
| | - Changhoon Choi
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Republic of Korea; (S.U.N.); (S.-W.S.); (Y.K.)
| | - Won Park
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Republic of Korea; (S.U.N.); (S.-W.S.); (Y.K.)
- Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| |
Collapse
|
22
|
Zhou L, Liu Y, Wu Y, Yang X, Spring Kong FM, Lu Y, Xue J. Low-dose radiation therapy mobilizes antitumor immunity: New findings and future perspectives. Int J Cancer 2024; 154:1143-1157. [PMID: 38059788 DOI: 10.1002/ijc.34801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023]
Abstract
Radiotherapy has unique immunostimulatory and immunosuppressive effects. Although high-dose radiotherapy has been found to have systemic antitumor effects, clinically significant abscopal effects were uncommon on the basis of irradiating single lesion. Low-dose radiation therapy (LDRT) emerges as a novel approach to enhance the antitumor immune response due to its role as a leverage to reshape the tumor immune microenvironment (TIME). In this article, from bench to bedside, we reviewed the possible immunomodulatory role of LDRT on TIME and systemic tumor immune environment, and outlined preclinical evidence and clinical application. We also discussed the current challenges when LDRT is used as a combination therapy, including the optimal dose, fraction, frequency, and combination of drugs. The advantage of low toxicity makes LDRT potential to be applied in multiple lesions to amplify antitumor immune response in polymetastatic disease, and its intersection with other disciplines might also make it a direction for radiotherapy-combined modalities.
Collapse
Affiliation(s)
- Laiyan Zhou
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Disaster Medical Center, Sichuan University, Chengdu, China
| | - Yuanxin Liu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanjun Wu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Yang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feng-Ming Spring Kong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
He X, Ji J, Qdaisat A, Esteva FJ, Yeung SCJ. Long-term overall survival of patients who undergo breast-conserving therapy or mastectomy for early operable HER2-Positive breast cancer after preoperative systemic therapy: an observational cohort study. LANCET REGIONAL HEALTH. AMERICAS 2024; 32:100712. [PMID: 38495316 PMCID: PMC10943473 DOI: 10.1016/j.lana.2024.100712] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 02/15/2024] [Accepted: 02/26/2024] [Indexed: 03/19/2024]
Abstract
Background Understanding the survival outcomes associated with breast-conserving therapy (BCT) and mastectomy after preoperative systemic therapy (PST) enables clinicians to provide more personalized treatment recommendations. However, lack of firm survival benefit data limits the breast surgery choices of human epidermal growth factor receptor 2 (HER2)-positive breast cancer patients who receive PST. We sought to determine whether BCT or mastectomy after PST for early operable HER2-positive breast cancer is associated with better long-term survival outcomes and determine the degree to which PST response affects this association. Methods In this observational cohort study, we compared the long-term survival outcomes of BCT and mastectomy after PST for HER2-positive breast cancer and evaluated the impact of PST response on the relationship between breast surgery performed and survival outcomes. Our cohort included 625 patients with early operable HER2-positive breast cancer who received PST followed by BCT or mastectomy between January 1998 and October 2009. These patients also received standard postoperative radiation, trastuzumab, and endocrine therapy as indicated clinically. We used propensity score matching to assemble mastectomy and BCT cohorts with similar baseline characteristics and used Kaplan-Meier plots and Cox proportional hazards regression to detect associations between surgery types and outcomes. Furthermore, in this study, we analyzed the original data of 625 patients using the inverse probability of treatment weighting (IPTW) method to enhance the reliability of the comparison between the mastectomy and BCT cohorts by addressing potential confounding variables. Findings Propensity score matching yielded cohorts of 221 patients who received BCT and 221 patients who underwent mastectomy. At the median follow-up time of 9.9 years, compared with BCT, mastectomy was associated with worse overall survival (hazard ratio, 1.66; 95% confidence interval [CI]: 1.08-2.57; P = 0.02). In patients who had axillary lymph node pathological complete response, mastectomy was associated with worse overall survival before matching (hazard ratio, 2.17; 95% CI: 1.22-3.86; P < 0.01) and after matching (hazard ratio, 2.12; 95% CI: 1.15-3.89; P = 0.02). Among patients with pathological complete response in the breast, the survival results did not differ significantly between BCT and mastectomy patients. IPTW method validated that BCT offers better overall survival in patients who had axillary lymph node pathological complete response. Interpretation People with HER2-positive breast cancer who have already had PST are more likely to survive after BCT, especially if they get a pathological complete response in the axillary lymph nodes. These findings underscore the necessity for further investigation into how responses to PST can inform the choice of surgical intervention and the potential impact on overall survival. Such insights could lead to the development of innovative tools that support personalized surgical strategies in the management of breast cancer. Funding This work was supported by grants from the Nantong Science and Technology Project (JCZ2022079), Nantong Health Commission Project (QA2021031, MSZ2023040) and National Natural Science Foundation of China (No. 82394430).
Collapse
Affiliation(s)
- Xuexin He
- Department of Medical Oncology, Huashan Hospital of Fudan University, Shanghai, China
- Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiali Ji
- Department of Medical Oncology, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Aiham Qdaisat
- Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francisco J. Esteva
- Division of Hematology/Oncology, Northwell Health Cancer Institute at Lenox Hill Hospital, New York, NY, USA
| | - Sai-Ching J. Yeung
- Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
24
|
Li J, Guo S, Li T, Hu S, Xu J, Xu X. Long non-coding RNA CCAT1 acts as an oncogene to promote radiation resistance in lung adenocarcinoma: an epigenomics-based investigation. Funct Integr Genomics 2024; 24:52. [PMID: 38448654 DOI: 10.1007/s10142-024-01330-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/17/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Long non-coding RNAs (lncRNAs) appear to be the crucial modulators in various processes and critically influence the oncogenesis. As one of the LncRNAs, LncRNA CCAT1 has been reported to be closely associated with the progression multiple cancers, but its role in modulating the radioresistance of lung adenocarcinoma (LUAD) remains unclear. In our present study, we screened the potential radioresistance related LncRNAs in LUAD based on the data from The Cancer Genome Atlas (TCGA) database. Data suggested that CCAT1 was abundantly expressed in LUAD and CCAT1 was significantly associated with poor prognosis and radioresistance. Moreover, our in vitro experiments showed that radiation treatment could trigger elevated expression of CCAT1 in the human LUAD cell lines. Further loss/gain-of-function investigations indicated that CCAT1 knockdown significantly inhibited cell proliferation, migration and promoted cell apoptosis in NCI-H1299 cells under irradiation, whereas CCAT1 overexpression in A549 cells yield the opposite effects. In summary, we identified the promoting role of CCAT1 in radioresistance of LUAD, which may provide a theoretical basis for radiotherapy sensitization of LUAD.
Collapse
Affiliation(s)
- Jian Li
- Department of Radiotherapy, Harbin Medical University Cancer Hospital, No.150 Haping Street, Harbin, 150076, Heilongjiang, China
| | - Shengnan Guo
- Department of Pathology, Harbin Medical University, Harbin, 150081, China
| | - Tianhao Li
- Department of Pathology, Harbin Medical University, Harbin, 150081, China
| | - Songliu Hu
- Department of Radiotherapy, Harbin Medical University Cancer Hospital, No.150 Haping Street, Harbin, 150076, Heilongjiang, China
| | - Jianyu Xu
- Department of Radiotherapy, Harbin Medical University Cancer Hospital, No.150 Haping Street, Harbin, 150076, Heilongjiang, China
| | - Xiangying Xu
- Department of Radiotherapy, Harbin Medical University Cancer Hospital, No.150 Haping Street, Harbin, 150076, Heilongjiang, China.
- Department of Radiotherapy, The Third Affilliated Hospital of Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
25
|
Qiu J, Xia Y, Bao Y, Cheng J, Liu L, Qian D. Silencing PinX1 enhances radiosensitivity and antitumor-immunity of radiotherapy in non-small cell lung cancer. J Transl Med 2024; 22:228. [PMID: 38431575 PMCID: PMC10908107 DOI: 10.1186/s12967-024-05023-y] [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: 12/01/2023] [Accepted: 02/23/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND We aimed to investigate the effects of PinX1 on non-small cell lung cancer(NSCLC) radiosensitivity and radiotherapy-associated tumor immune microenvironment and its mechanisms. METHODS The effect of PinX1 silencing on radiosensitivity in NSCLC was assessed by colony formation and CCK8 assay, immunofluorescence detection of γ- H2AX and micronucleus assay. Western blot was used to assess the effect of PinX1 silencing on DNA damage repair pathway and cGAS-STING pathway. The nude mouse and Lewis lung cancer mouse model were used to assess the combined efficacy of PinX1 silencing and radiotherapy in vivo. Changes in the tumor immune microenvironment were assessed by flow cytometry for different treatment modalities in the Lewis luuse model. The interaction protein RBM10 was screened by immunoprecipitation-mass spectrometry. RESULTS Silencing PinX1 enhanced radiosensitivity and activation of the cGAS-STING pathway while attenuating the DNA damage repair pathway. Silencing PinX1 further increases radiotherapy-stimulated CD8+ T cell infiltration and activation, enhances tumor control and improves survival in vivo; Moreover, PinX1 downregulation improves the anti-tumor efficacy of radioimmunotherapy, increases radioimmune-stimulated CD8+ T cell infiltration, and reprograms M2-type macrophages into M1-type macrophages in tumor tissues. The interaction of PinX1 and RBM10 may promote telomere maintenance by assisting telomerase localization to telomeres, thereby inhibiting the immunostimulatory effects of IR. CONCLUSIONS In NSCLC, silencing PinX1 significantly contributed to the radiosensitivity and promoted the efficacy of radioimmunotherapy. Mechanistically, PinX1 may regulate the transport of telomerase to telomeres through interacting with RBM10, which promotes telomere maintenance and DNA stabilization. Our findings reveal that PinX1 is a potential target to enhance the efficacy of radioimmunotherapy in NSCLC patients.
Collapse
Affiliation(s)
- Jieping Qiu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ying Xia
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yawei Bao
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jingjing Cheng
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Lei Liu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Dong Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
26
|
Huang P, Ning X, Kang M, Wang R. Ferroptosis-Related Genes Are Associated with Radioresistance and Immune Suppression in Head and Neck Cancer. Genet Test Mol Biomarkers 2024; 28:100-113. [PMID: 38478802 PMCID: PMC10979683 DOI: 10.1089/gtmb.2023.0193] [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] [Indexed: 04/01/2024] Open
Abstract
Background: Ferroptosis is associated with tumor development; however, its contribution to radioresistant head and neck cancer (HNC) remains unclear. In this study, we used bioinformatics analysis and in vitro testing to explore ferroptosis-related genes associated with HNCs radiosensitivity. Materials and Methods: GSE9714, GSE90761, and The Cancer Genome Atlas (TCGA) datasets were searched to identify ferroptosis-related differentially expressed genes between radioresistant and radiosensitive HNCs or radiation-treated and nonradiation-treated HNCs. A protein-protein interaction analysis on identified hub genes was then performed. Receiver operating characteristic curves and Kaplan-Meier survival analysis were used to assess the diagnostic and prognostic potential of the hub genes. Cell counting kit-8, transwell assay, and flow cytometry were applied to examine the role of hub gene collagen type IV, alpha1 chain (COL4A1) on the proliferation, migration, invasion, and apoptosis of TU686 cells. Results: Hub genes MMP10, MMP1, COL4A1, IFI27, and INHBA showed diagnostic potential for HNC and were negatively correlated with overall survival and disease-free survival in the TCGA dataset. Also, IL-1B, IFI27, INHBA, and COL4A1 mRNA levels were significantly increased in TCGA patients with advanced clinical stages or receiving radiotherapy, whereas COL4A1, MMP10, and INHBA expressions were negatively correlated with immune infiltration. Furthermore, the knockdown of COL4A1 inhibited cell proliferation, migration, and invasion while promoting apoptosis in TU686 cells. Conclusion: Ferroptosis-related hub genes, such as COL4A1, are potential diagnostic and prognostic indicators as well as therapeutic targets for HNC.
Collapse
Affiliation(s)
- Ping Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Oncology, LiuZhou Traditional Chinese Medical Hospital Affiliated to Guangxi University of Chinese Medicine, Liuzhou, China
| | - Xuejian Ning
- Department of Oncology, LiuZhou Traditional Chinese Medical Hospital Affiliated to Guangxi University of Chinese Medicine, Liuzhou, China
| | - Min Kang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - RenSheng Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
27
|
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
|
28
|
Zhong M, Fang Z, Guo W, Yu X. Translation regulatory long non-coding RNA 1 negatively regulates cell radiosensitivity via the miR-22-3p/SP1 axis in non-small cell lung cancer. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e13734. [PMID: 38286742 PMCID: PMC10824624 DOI: 10.1111/crj.13734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 10/11/2023] [Accepted: 01/05/2024] [Indexed: 01/31/2024]
Abstract
OBJECTIVE Non-small cell lung cancer (NSCLC) occupies 85% of lung cancer. Long non-coding RNAs (LncRNAs) can regulate the radiosensitivity of cancers. This study explored the mechanism of lncRNA TRERNA1 in the radiosensitivity of NSCLC cells. METHODS LncRNA TRERNA1 level in NSCLC cell lines was determined. NSCLC cell radiation tolerance was measured. TRERNA1 expression was silenced or overexpressed in A549/HCC827 cells with the highest/lowest radiation tolerance, respectively. The contents of γ-H2AX and SA-β-gal in NSCLC cells after radiation induction were detected. The targeted binding of TRERNA1 to miR-22-3p and miR-22-3p to SP1 were verified by dual-luciferase assay. SP1 expression were detected. Functional rescue experiments were implemented to confirm the roles of miR-22-3p and SP1 in the regulatory mechanism of TRERNA1. RESULTS TRERNA1 was upregulated in NSCLC cells. TRERNA1 silencing enhanced radiosensitivity of NSCLC cells. TRERNA1 silencing elevated the contents of γ-H2AX and SA-β-gal in A549 cells after radiation induction, while TRERNA1 overexpression showed an opposite trend in HCC827 cells. There were targeting relationships between TRERNA1 and miR-22-3p, and miR-22-3p and SP1. miR-22-3p repression or SP1 overexpression abolished the effects of TRERNA1 silencing. CONCLUSION TRERNA1 silencing enhanced radiosensitivity of NSCLC cells via the miR-22-3p/SP1 axis. This study may offer new targets for NSCLC treatment.
Collapse
Affiliation(s)
- Ming Zhong
- Department of Thoracic SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamenChina
| | - Zheng Fang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamenChina
| | - Weixi Guo
- Department of Thoracic SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamenChina
| | - Xiuyi Yu
- Department of Thoracic SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamenChina
| |
Collapse
|
29
|
Song KH, Jung SY, Park JI, Lee DH, Ahn J, Hwang SG, Lim DS, Song JY. Poliovirus receptor inhibition in breast cancer cells induces antitumor immunity via T cell activation. Am J Cancer Res 2023; 13:5966-5980. [PMID: 38187056 PMCID: PMC10767338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/02/2023] [Indexed: 01/09/2024] Open
Abstract
Radiotherapy (RT) is a commonly used treatment option for patients with cancer because it can effectively control tumor growth and kill tumor cells. However, the impact of RT goes beyond direct tumor cell killing because it can change the tumor microenvironment by altering surrounding tissues and infiltrating cells and modulating the expression of immune checkpoints. Poliovirus receptor (PVR, cluster of differentiation (CD)155), a member of the nectin-like molecule family, is overexpressed in many human cancers. However, its role in the tumor growth and T-cell immune responses of triple-negative breast cancer (TNBC) remains unclear. In the present study, we observe that radiation exposure increases PVR expression in MDA-MB-231 and BT549 cells. Silencing PVR not only inhibited the proliferation of breast cancer cells but also significantly enhanced the cytotoxicity of cytotoxic T lymphocytes (CTLs) compared with the control or RT groups. Treatment of T cells with PVR decreased CD8+ T cells, increased CD4+ T cells, and induced PVR ligands such as T cell immunoreceptor with immunoglobulin and ITIM domain, CD226, and CD96. However, after treatment with PVR, CTL responses decreased and secretion of interferon-γ, tumor necrosis factor-α, interleukin (IL)-2, IL-6, and IL-10 was significantly inhibited. In contrast, PVR knockdown increased the production of these cytokines, illustrating the immunosuppressive function of PVR. Suppression of PVR using an anti-PVR antibody inhibited 4T1 tumor growth by increasing immune cell infiltration. These results provide new insights into the role of PVR in TNBC and highlight its potential as a target for T cell-mediated immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Kyung-Hee Song
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| | - Seung-Youn Jung
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| | - Jeong-In Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| | - Dong-Hyeon Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| | - Jiyeon Ahn
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| | - Sang-Gu Hwang
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| | - Dae-Seog Lim
- Department of Biotechnology, CHA UniversityGyeonggi-do 13488, Republic of Korea
| | - Jie-Young Song
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| |
Collapse
|
30
|
Zhang Z, Peng Y, Peng X, Xiao D, Shi Y, Tao Y. Effects of radiation therapy on tumor microenvironment: an updated review. Chin Med J (Engl) 2023; 136:2802-2811. [PMID: 37442768 PMCID: PMC10686612 DOI: 10.1097/cm9.0000000000002535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Indexed: 07/15/2023] Open
Abstract
ABSTRACT Cancer is a major threat to human health and causes death worldwide. Research on the role of radiotherapy (RT) in the treatment of cancer is progressing; however, RT not only causes fatal DNA damage to tumor cells, but also affects the interactions between tumor cells and different components of the tumor microenvironment (TME), including immune cells, fibroblasts, macrophages, extracellular matrix, and some soluble products. Some cancer cells can survive radiation and have shown strong resistance to radiation through interaction with the TME. Currently, the complex relationships between the tumor cells and cellular components that play major roles in various TMEs are poorly understood. This review explores the relationship between RT and cell-cell communication in the TME from the perspective of immunity and hypoxia and aims to identify new RT biomarkers and treatment methods in lung cancer to improve the current status of unstable RT effect and provide a theoretical basis for further lung cancer RT sensitization research in the future.
Collapse
Affiliation(s)
- Zewen Zhang
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan 410078, China
| | - Yuanhao Peng
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xin Peng
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Desheng Xiao
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ying Shi
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan 410078, China
| | - Yongguang Tao
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
31
|
Najafi A, Keykhaee M, Kazemi MH, Karimi MY, Khorramdelazad H, Aghamohamadi N, Bolouri MR, Ghaffari-Nazari H, Mirsharif ES, Karimi M, Dehghan Manshadi HR, Mahdavi SR, Safari E, Jalali SA, Falak R, Khoobi M. Catalase-gold nanoaggregates manipulate the tumor microenvironment and enhance the effect of low-dose radiation therapy by reducing hypoxia. Biomed Pharmacother 2023; 167:115557. [PMID: 37757491 DOI: 10.1016/j.biopha.2023.115557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Radiotherapy as a standard method for cancer treatment faces tumor recurrence and antitumoral unresponsiveness. Suppressive tumor microenvironment (TME) and hypoxia are significant challenges affecting efficacy of radiotherapy. Herein, a versatile method is introduced for the preparation of pH-sensitive catalase-gold cross-linked nanoaggregate (Au@CAT) having acceptable stability and selective activity in tumor microenvironment. Combining Au@CAT with low-dose radiotherapy enhanced radiotherapy effects via polarizing protumoral immune cells to the antitumoral landscape. This therapeutic approach also attenuated hypoxia, confirmed by downregulating hypoxia hallmarks, such as hypoxia-inducible factor α-subunits (HIF-α), vascular endothelial growth factor (VEGF), and EGF. Catalase stability against protease digestion was improved significantly in Au@CAT compared to the free catalase. Moreover, minimal toxicity of Au@CAT on normal cells and increased reactive oxygen species (ROS) were confirmed in vitro compared with radiotherapy. Using the nanoaggregates combined with radiotherapy led to a significant reduction of immunosuppressive infiltrating cells such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (T-regs) compared to the other groups. While, this combined therapy could significantly increase the frequency of CD8+ cells as well as M1 to M2 macrophages (MQs) ratio. The combination therapy also reduced the tumor size and increased survival rate in mice models of colorectal cancer (CRC). Our results indicate that this innovative nanocomposite could be an excellent system for catalase delivery, manipulating the TME and providing a potential therapeutic strategy for treating CRC.
Collapse
Affiliation(s)
- Alireza Najafi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Keykhaee
- Department of Pharmaceutical Biomaterials and Medical Biomaterial Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Kazemi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Khorramdelazad
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Nazanin Aghamohamadi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Bolouri
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haniyeh Ghaffari-Nazari
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Milad Karimi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Seied Rabi Mahdavi
- Radiation Biology Research Center& Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Safari
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Jalali
- Immunology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mehdi Khoobi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Sun L, Zhou H, Wu C, Peng Y. Molecular markers that predict response to combined radiotherapy and immunotherapy in patients with lung adenocarcinoma: a bioinformatics analysis. Transl Cancer Res 2023; 12:2646-2659. [PMID: 37969379 PMCID: PMC10643968 DOI: 10.21037/tcr-23-968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/19/2023] [Indexed: 11/17/2023]
Abstract
Background Immunotherapy has had a high success rate in treating lung adenocarcinoma (LUAD) for several decades. However, many patients do not benefit from immunotherapy alone. Recent studies revealed that a combination of immunotherapy and radiotherapy (RT) stimulates a good systemic immune response to LUAD. However, clinical and experimental evidence suggest that RT may give rise to primary immunodeficiency, facilitating tumor immunity escape. Little is known about the molecular mechanisms whereby RT and stereotactic body radiotherapy (SBRT) influence tumor immunogenicity and the effectiveness of immunotherapy in patients with LUAD. Methods We investigated molecular markers that predict response to combination of immunotherapy and SBRT in the treatment of LUAD using bioinformatics. Results SBRT significantly upregulated the expression of PTPRC, LILRB2, TLR8, CCR5, and PLEK and significantly downregulated the expression of CXCL13, CD19, and LTA. Among these genes, the expression of PTPRC, TLR8, and CCR5 was associated with responsiveness to immunotherapy after SBRT. However, only TLR8 and CCR5 expression were associated with an improved prognosis. Further analysis revealed that TLR8 and CCR5 expression increased responsiveness to immunotherapy by promoting M0 macrophage and memory B cell infiltration of LUAD tissues. Conclusions In patients with LUAD, TLR8 and CCR5 expression are potential markers of a favorable response to combined immunotherapy and RT.
Collapse
Affiliation(s)
- Lu Sun
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiting Zhou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Wu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Yi Peng
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Kong Y, Xie F, Zhang Z, Wang S, Zhang Y, Di Y, Zhou Z, Jiang D, Li J, Huang Q, Wang J, Li X, Pan Z, Ni R, Guan Y. Evaluation of novel anti-CEACAM6 antibody-based conjugates for radioimmunotheranostics of pancreatic ductal adenocarcinoma. Eur Radiol 2023; 33:7077-7088. [PMID: 37166496 DOI: 10.1007/s00330-023-09679-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 01/13/2023] [Accepted: 02/22/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant solid tumor that lacks early diagnostic methods. Recently, targeted immunotherapy and radiotherapy have been integrated with radionuclide-antibody conjugate drugs, which can be used for targeted diagnosis and dynamic imaging of tumors. CEACAM6 is overexpressed in pancreatic tumors and is a potential theranostic target for PDAC. We aimed to develop a novel targeted carrier for theranostics of PDAC and other solid tumors. METHODS Based on camelid heavy-chain-only antibodies, we developed a CEACAM6-targeting recombinant antibody NY004, and evaluated it as a novel antibody-carrier for imaging and therapy of cancer in tumor models. We labeled NY004 with theranostic nuclides and applied this self-developed antibody platform in diagnostic imaging and antitumor assessment in PDAC models. RESULTS Through microPET, IHC, and biodistribution assays, targeting and biodistribution of [89Zr]-NY004 in solid tumors including PDAC was examined, and the investigated tumors were all CEACAM6-positive malignancies. We found that NY004 was suitable for use as a drug carrier for radioimmunotheranostics. Our study showed that NY004 was characterized by high targeted uptake and a long retention time in PANC-1 tumors (up to 6 days post-injection), with good specificity and high imaging efficiency. Therapeutic evaluation of the radionuclide-labeled antibody drug [177Lu]-NY004 in PDAC tumor-bearing model revealed that NY004 had high and prolonged uptake in tumors, relatively low non-target organ uptake, and good anti-tumor efficacy. CONCLUSION As a drug platform for radiotheranostics, CEACAM6-specific antibody NY004 met the requirements of easy-labeling, targeting specificity, and effective persistence in pancreatic adenocarcinoma tissues. KEY POINTS • [89Zr]-NY004 has good specificity and high imaging efficiency, and is characterized by high tumor-targeting uptake and a long tumor retention time as a PET molecular imaging tracer. • Therapeutic radionuclide-conjugated antibody drug [177Lu]-NY004 has high uptake and prolonged uptake duration in tumors, low non-target organ uptake, and significant tumor-inhibiting efficacy in PDAC model. • The self-developed antibody structure NY004 is a promising drug platform for radioimmunotheranostics of CEACAM6-positive tumors including pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China.
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China
| | - Zhengwei Zhang
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China
| | - Shaobo Wang
- PET/CT Center, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China
| | - Yabin Zhang
- Department of Nuclear Medicine, Third People's Hospital of Honghe State, Honghe, 661000, Yunnan, China
| | - Yang Di
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zhongwen Zhou
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Donglang Jiang
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China
| | - Junpeng Li
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China
| | - Jie Wang
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China
| | - Xiuming Li
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China
| | - Zhiwei Pan
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China
| | - Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich & University of Zurich, Zurich, Switzerland.
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China.
| |
Collapse
|
34
|
Cheng F, Su T, Zhou S, Liu X, Yang S, Lin S, Guo W, Zhu G. Single-dose injectable nanovaccine-in-hydrogel for robust immunotherapy of large tumors with abscopal effect. SCIENCE ADVANCES 2023; 9:eade6257. [PMID: 37450588 PMCID: PMC10348685 DOI: 10.1126/sciadv.ade6257] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 06/09/2023] [Indexed: 07/18/2023]
Abstract
Current cancer immunotherapy [e.g., immune checkpoint blockade (ICB)] only benefits small subsets of patients, largely due to immunosuppressive tumor microenvironment (TME). In situ tumor vaccination can reduce TME immunosuppression and thereby improve cancer immunotherapy. Here, we present single-dose injectable (nanovaccines + ICBs)-in-hydrogel (NvIH) for robust immunotherapy of large tumors with abscopal effect. NvIH is thermo-responsive hydrogel co-encapsulated with ICB antibodies and novel polymeric nanoparticles loaded with three immunostimulatory agonists for Toll-like receptors 7/8/9 (TLR7/8/9) and stimulator of interferon genes (STING). Upon in situ tumor vaccination, NvIH undergoes rapid sol-to-gel transformation, prolongs tumor retention, sustains the release of immunotherapeutics, and reduces acute systemic inflammation. In multiple poorly immunogenic tumor models, single-dose NvIH reduces multitier TME immunosuppression, elicits potent TME and systemic innate and adaptive antitumor immunity with memory, and regresses both local (vaccinated) and distant large tumors with abscopal effect, including distant orthotopic glioblastoma. Overall, NvIH holds great potential for tumor immunotherapy.
Collapse
Affiliation(s)
- Furong Cheng
- Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy, The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Ting Su
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy, The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Shurong Zhou
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy, The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiang Liu
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy, The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Suling Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shuibin Lin
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Weisheng Guo
- Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Guizhi Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
35
|
Zhou Z, Guan B, Xia H, Zheng R, Xu B. Particle radiotherapy in the era of radioimmunotherapy. Cancer Lett 2023:216268. [PMID: 37331583 DOI: 10.1016/j.canlet.2023.216268] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/24/2023] [Accepted: 06/11/2023] [Indexed: 06/20/2023]
Abstract
Radiotherapy (RT) is one of the key modalities for cancer treatment, and more than 70% of tumor patients will receive RT during the course of their disease. Particle radiotherapy, such as proton radiotherapy, carbon-ion radiotherapy (CIRT) and boron neutron capture therapy (BNCT), is currently available for the treatment of patients Immunotherapy combined with photon RT has been successfully used in the clinic. The effect of immunotherapy combined with particle RT is an area of interest. However, the molecular mechanisms underlying the effects of combined immunotherapy and particle RT remain largely unknown. In this review, we summarize the properties of different types of particle RT and the mechanisms underlying their radiobiological effects. Additionally, we compared the main molecular players in photon RT and particle RT and the mechanisms involved the RT-mediated immune response.
Collapse
Affiliation(s)
- Zihan Zhou
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Bingjie Guan
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Huang Xia
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Rong Zheng
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China; Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China.
| | - Benhua Xu
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China; Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China.
| |
Collapse
|
36
|
Xian F, Wu J, Zhong L, Xu G. Efficacy and safety of PD1/PDL1 inhibitors combined with radiotherapy and anti-angiogenic therapy for solid tumors: A systematic review and meta-analysis. Medicine (Baltimore) 2023; 102:e33204. [PMID: 36897735 PMCID: PMC9997836 DOI: 10.1097/md.0000000000033204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/15/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND The triple combination of programmed cell death 1 (PD1)/programmed cell death ligand 1 (PDL1) inhibitors, radiotherapy (RT), and anti-angiogenesis agents has been widely used in the treatment of solid tumors and has shown positive efficacy. We conducted a meta-analysis to evaluate the efficacy and safety of PD1/PDL1 inhibitors combined with anti-angiogenic agents and RT for the treatment of solid cancers. METHODS A systematic search of PubMed, Embase, Cochrane Library, and Web of Science databases was conducted from inception to October 31, 2022. Studies involving patients with solid cancers who received PD1/PDL1 inhibitors combined with RT and anti-angiogenic agents treatment that reported overall response rate, complete remission rate, disease control rate, and adverse events (AEs) were included. A random-effects or fixed-effects model was used for the pooled rates, and 95% confidence intervals (CIs) were determined for all outcomes. The quality of the included literature was assessed using the methodological index for nonrandomized studies critical appraisal checklist. Egger test was used to assess the publication bias in the included studies. RESULTS Ten studies (4 nonrandomized controlled trials and 6 single-arm trials), including 365 patients, were identified and included in the meta-analysis. The pooled overall response rate after treatment with PD1/PDL1 inhibitors combined with RT and anti-angiogenic agents was 59% (95% CI: 48-70%), whereas the disease control rate and complete remission rate were 92% (95% CI: 81-103%) and 48% (95% CI: 35-61%), respectively. Moreover, the meta-analysis showed that compared with triple-regimen, monotherapy or dual-combination treatment did not improve overall survival (hazard ratio = 0.499, 95% CI: 0.399-0.734) and progression-free survival (hazard ratio = 0.522, 95% CI: 0.352-0.774). The pooled rate of grade 3 to 4 AEs was 26.9% (95% CI: 7.8%-45.9), and the common AEs to triple therapy included leukopenia (25%), thrombocytopenia (23.8%), fatigue (23.2%), gastrointestinal discomfort (22%), increased alanine aminotransferase (22%), and neutropenia (21.4%). CONCLUSION In the treatment of solid tumors, PD1/PDL1 inhibitors combined with RT and anti-angiogenic drugs achieved a positive response and better survival benefits than monotherapy or dual therapy. In addition, combination therapy is tolerable and safe. REGISTRATION PROSPERO ID: CRD42022371433.
Collapse
Affiliation(s)
- Feng Xian
- Department of Oncology, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Wu
- Department of Operations Management, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, China
| | - Liming Zhong
- Department of Operations Management, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, China
| | - Guohui Xu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
37
|
Kang P, Liu D, Li L, Guo X, Ye Y, Li Y, Jiang Q, Lin S, Yuan Q. Interleukin 8 in plasma is an efficacy marker for advanced non-small cell lung cancer treated with hypofractionated radiotherapy and PD-1 blockade. Cytokine 2023; 163:156133. [PMID: 36724715 DOI: 10.1016/j.cyto.2023.156133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 01/31/2023]
Abstract
BACKGROUND Programmed death-1 (PD-1) blockade promotes combination therapy in advanced non-small cell lung cancer (NSCLC), hypofractionated radiotherapy (HFRT) and chemotherapy combined with immunotherapy improves the outcome of prognosis in advanced NSCLC, while effective biomarkers to follow prognostic efficacy are still to be found. METHODS We enrolled 44 NSCLC patients with HFRT combined with PD-1 blockade, 13 patients with chemotherapy combined with immunotherapy, additionally collected tissue samples from 8 patients with earlystage NSCLC without therapy, and peripheral whole blood from 16 healthy donors, detected the expression differences of cytokines Interleukin 6 (IL-6), Interleukin 8 (IL-8) and Interleukin 17A (IL-17A) in the peripheral plasma and tissues by flow cytometry, immunofluorescence, and real-time fluorescence quantitative PCR. Cultured peripheral blood mononuclear cell (PBMC) and tumor-infiltrating T cells with recombinant human IL-8 in vitro to observe the changes of immune memory T cell subtypes and apoptosis. RESULTS Our results show that IL-6, IL-8, and IL-17A are highly expressed in advanced NSCLC, high levels of IL-8 are significantly associated with poor prognosis in advanced NSCLC patients treated with HFRT + PD1 blockade, high circulating IL-8 in NSCLC increased apoptosis of effector memory RA (TemRA; CD45RA+CCR7-) T cell subsets and CD8+ T cell subsets in tissues, resulting in decreased peripheral TemRA and stem cell-like memory T cells (TSCM: CD45RA +CCR7 + CD95 +) in tissue. CONCLUSION We suggest that IL-8 can impair immune memory function in NSCLC. It is a useful biomarker to evaluate the efficacy of HFRT + PD1 blockade in advanced NSCLC. Further exploration of easily available plasma biomarkers for personalized treatment of NSCLC is required.
Collapse
Affiliation(s)
- Pengyuan Kang
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Dan Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Lin Li
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Xiyuan Guo
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Yingchun Ye
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Yunfei Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qin Jiang
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Sheng Lin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| | - Qing Yuan
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; Institute of Nuclear Medicine, Southwest Medical University, Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, China.
| |
Collapse
|
38
|
Wang Q, Shao X, Zhang Y, Zhu M, Wang FXC, Mu J, Li J, Yao H, Chen K. Role of tumor microenvironment in cancer progression and therapeutic strategy. Cancer Med 2023. [PMID: 36807772 DOI: 10.1002/cam4.5698] [Citation(s) in RCA: 104] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/18/2023] [Accepted: 02/02/2023] [Indexed: 02/23/2023] Open
Abstract
Cancer is now considered a tumor microenvironment (TME) disease, although it was originally thought to be a cell and gene expression disorder. Over the past 20 years, significant advances have been made in understanding the complexity of the TME and its impact on responses to various anticancer therapies, including immunotherapies. Cancer immunotherapy can recognize and kill cancer cells by regulating the body's immune system. It has achieved good therapeutic effects in various solid tumors and hematological malignancies. Recently, blocking of programmed death-1 (PD-1), programmed death-1 ligand-1 (PD-L1), and programmed death Ligand-2 (PD-L2), the construction of antigen chimeric T cells (CAR-T) and tumor vaccines have become popular immunotherapies Tumorigenesis, progression, and metastasis are closely related to TME. Therefore, we review the characteristics of various cells and molecules in the TME, the interaction between PD-1 and TME, and promising cancer immunotherapy therapeutics.
Collapse
Affiliation(s)
- Qingjing Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Xueting Shao
- Institute of Pharmaceutical Biotechnology & Research Center for Clinical Pharmacy, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuxuan Zhang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Miaojin Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Frederick X C Wang
- The EnMed Program at Houston Methodist Hospital, Texas A&M University College of Medicine and College of Engineering, Houston, Texas, USA
| | - Jianjian Mu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jiaxuan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Hangping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| |
Collapse
|
39
|
Yang L, Zhao Q, Wang X, Pilapong C, Li Y, Zou J, Jin J, Rong J. Investigation on the regulatory T cells signature and relevant Foxp3/STAT3 axis in esophageal cancer. Cancer Med 2023; 12:4993-5008. [PMID: 36226375 PMCID: PMC9972178 DOI: 10.1002/cam4.5194] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 08/05/2022] [Accepted: 08/14/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) have an important role in accelerating the immunosuppression of tumor. Tregs regulation is a hopeful strategy to improve the dismal prognosis of Esophageal cancer (EC), while its mechanisms have not yet been fully clarified. METHODS To characterize the role of Tregs in EC, we comprehensively explored its prognostic value, clinical pathology partnership, related biological functions and potential mechanisms at transcriptome level. Through the integrated analysis of GEO and TCGA datasets, we comprehensively evaluated the Tregs infiltration patterns in EC patients. The correlation between Tregs infiltration and genomic characteristics, as well as biological functions were analyzed by a variety of computational algorithms. RESULTS We observed that Tregs were significantly upregulated in EC and involved in various immune processes. According to TCGA and GEO transcriptional classification schemes, Tregs specific genes were observed to be highly expressed in tumor samples, as well as were closely associated with poor prognosis and worse clinical outcomes. In addition, EC patients can be stratified into high-risk and low-risk immune subgroups according to Tregs/macrophages infiltration level, and the results showed significant differences in tumor development, biological processes and probe gene expression pattern. The multi-variate analysis revealed that the interaction between STAT3 and Foxp3 was a potential prognostic signature of Tregs in EC, especially the modulation effect of STAT3 on Foxp3 expression, which has not been well studied in EC. We also identified that STAT3 and Foxp3 expression presented a high accuracy in predicting Tregs infiltration level in EC patients (AUC: 0.817; 95% CI: 0.756-0.878). CONCLUSIONS Our results revealed that Tregs have the potential to predict prognosis and tumor deterioration in EC patients. A comprehensive landscape of Tregs regulation mechanisms will help us interpret the immunosuppression of tumor microenvironment (TME) and novel strategies for EC immunotherapy.
Collapse
Affiliation(s)
- Lin Yang
- Department of Oncology, The Second People's Hospital of Yibin, Yibin, People's Republic of China.,Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qijie Zhao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, People's Republic of China.,Center of Excellence for Molecular Imaging (CEMI), Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Xing Wang
- Shichuan Nursing Vocational College, Chengdu, People's Republic of China
| | - Chalermchai Pilapong
- Center of Excellence for Molecular Imaging (CEMI), Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Yi Li
- Department of Oncology, The Second People's Hospital of Yibin, Yibin, People's Republic of China
| | - Jun Zou
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jing Jin
- Department of Oncology, The Second People's Hospital of Yibin, Yibin, People's Republic of China
| | - Jinfeng Rong
- Department of Oncology, The Second People's Hospital of Yibin, Yibin, People's Republic of China
| |
Collapse
|
40
|
Chen N, Wang Y, Zeng Y, Li Y, Pan Z, Li H, Chen J, Chen Z, Yuan J, Yan W, Lu YJ, Liu X, He Y, Zhang K. All-in-one CoFe 2O 4@Tf nanoagent with GSH depletion and tumor-targeted ability for mutually enhanced chemodynamic/photothermal synergistic therapy. Biomater Sci 2023; 11:828-839. [PMID: 36453535 DOI: 10.1039/d2bm01542k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In the complex and severe tumor microenvironment, the antitumor efficiency of nanomedicines is significantly limited by their low-efficacy monotherapy, non-tumor targeting, and systemic toxicity. Herein, to achieve tumor-targeted and enhanced chemodynamic/photothermal therapy (CDT/PTT), we fabricated an "all-in-one" biocompatible transferrin-loaded cobalt ferrate nanoparticle (CoFe2O4@Tf (CFOT)) with multiple functions by a simple solvothermal method and the following transferrin (Tf) functionalization. Upon exposure to 808 nm laser irradiation, CFOT, as a novel photothermal agent, exhibited outstanding phototherapeutic activity because of its excellent photothermal conversion efficiency (η = 46.5%) for high-performance PTT. Moreover, CFOT with multiple redox pairs could efficiently convert endogenous H2O2 to hazardous hydroxyl radicals (˙OH) via Fenton reactions while scavenging overexpressed GSH in the tumor microenvironment to realize self-reinforcing CDT. Importantly, CFOT undergoes a promoted Fenton-type reaction upon increasing the temperature under a photothermal effect and could augment PTT by high-level ˙OH, exhibiting a considerably enhanced synergistic therapeutic effect. In vitro and in vivo experimental results demonstrated that CFOT has good potential as an "all-in-one" nanoagent to combine photothermal, chemodynamic, and tumor targeting for efficient tumor elimination.
Collapse
Affiliation(s)
- Niping Chen
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Yakun Wang
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Yaoxun Zeng
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Yushan Li
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Zhenxing Pan
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Haihong Li
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Jingman Chen
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Zefeng Chen
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Jiongpeng Yuan
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Wen Yan
- Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Yu-Jing Lu
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Xujie Liu
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Yan He
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Kun Zhang
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China. .,School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| |
Collapse
|
41
|
CD96 as a Potential Immune Regulator in Cancers. Int J Mol Sci 2023; 24:ijms24021303. [PMID: 36674817 PMCID: PMC9866520 DOI: 10.3390/ijms24021303] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/12/2023] Open
Abstract
The discovery of CTLA-4 and PD-1 checkpoints has prompted scientific researchers and the pharmaceutical industry to develop and conduct extensive research on tumor-specific inhibitors. As a result, the list of potential immune checkpoint molecules is growing over time. Receptors for nectin and nectin-like proteins have recently emerged as promising targets for cancer immunotherapy. Potential immune checkpoints, including CD226, TIGIT, and CD96, belong to this receptor class. Among them, CD96 has received little attention. In this mini-review, we aim to discuss the basic biology of CD96 as well as the most recent relevant research on this as a promising candidate for cancer immunotherapy.
Collapse
|
42
|
Gan C, Li M, Lu Y, Peng G, Li W, Wang H, Peng Y, Hu Q, Wei W, Wang F, Liu L, Zhao Q. SPOCK1 and POSTN are valuable prognostic biomarkers and correlate with tumor immune infiltrates in colorectal cancer. BMC Gastroenterol 2023; 23:4. [PMID: 36611136 PMCID: PMC9826581 DOI: 10.1186/s12876-022-02621-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 12/15/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Immune cells and stromal cells in the tumor microenvironment play a vital role in the progression of colorectal cancer (CRC). The study aimed to screen valuable prognostic biomarkers in CRC based on stromal and immune scores. METHOD The ESTIMATE algorithm was used to calculate the immune and stromal scores of CRC samples in TCGA. Then samples were divided into high and low score groups based on the median value of the scores. Differentially expressed genes (DEGs) associated with immune and stromal scores were screened. WGCNA and univariate COX regression analysis were performed to further identify key prognostic genes. Analysis of scRNA-seq for CRC was used for verifying the main source of the key genes. The prognostic value of they was validated based on The Gene Expression Profiling Interactive Analysis and GSE17536 dataset. TIMER and CIBERSORT algorithms were applied to analyze the correlations among key genes and tumor-infiltrating immune cells. Several pairs of colon cancer tissue were used to be proven. RESULT 1314 upregulated and 4 downregulated genes were identified, which were significantly enriched in immune-related biological processes and pathways. Among these DEGs, SPOCK1 and POSTN were identified as key prognostic genes and mainly expressed in cancer-associated fibroblasts for CRC. High expression of SPCOK1 and POSTN was associated with advanced clinical stage, T stage, N stage, and poor prognosis of CRC. The results from CIBERSORT and TIMER revealed that SPOCK1 and POSTN were associated with tumor-infiltrating immune cells, especially macrophages and neutrophils. Meanwhile, in several pairs of human colorectal tissue samples, SPOK1 and POSTN were found to be significantly overexpressed in colorectal tissue compared with para-cancer tissue, and macrophage surface markers CD68 (co-expressed by M1 and M2 macrophages) and CD206 (M2-specific macrophage expression) were also overexpressed in cancer tissue. Besides, SPOCK1 and POSTN expression were positively correlated with the expression of immune checkpoints. CONCLUSION Collectively, our results indicate that SPOCK1 and POSTN associated with CAF may be novel prognostic biomarkers in CRC and correlate with immune infiltrates.
Collapse
Affiliation(s)
- Caiqin Gan
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| | - Mengting Li
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| | - Yuanyuan Lu
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| | - Ganjing Peng
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| | - Wenjie Li
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| | - Haizhou Wang
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| | - Yanan Peng
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| | - Qian Hu
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| | - Wanhui Wei
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| | - Fan Wang
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| | - Lan Liu
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| | - Qiu Zhao
- grid.413247.70000 0004 1808 0969Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000 China ,grid.413247.70000 0004 1808 0969Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000 China
| |
Collapse
|
43
|
Zhang Y, Ding Y, Li N, Wang S, Zhou S, Li R, Yang H, Li W, Qu J. Noninvasive Imaging of Tumor PD-L1 Expression Using [ 99mTc]Tc-Labeled KN035 with SPECT/CT. Mol Pharm 2023; 20:690-700. [PMID: 36541699 DOI: 10.1021/acs.molpharmaceut.2c00874] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Programmed cell death protein-1/ligand-1 (PD-1/PD-L1) checkpoint blockade is a major breakthrough in cancer therapy, but identifying patients likely to benefit from this therapy remains challenging. Immunohistochemistry is not informative about PD-L1 expression heterogeneity because of the limitations of invasive tissue collection. Noninvasive SPECT imaging is an approach to patient selection and therapeutic monitoring by assessing the PD-L1 status throughout the whole body. Here, we radiolabeled a single-domain PD-L1 antibody with technetium-99m (99mTc) for immune-SPECT imaging to evaluate its feasibility of detecting PD-L1 expression. The radiochemical purity of [99mTc]Tc-HYNIC-KN035 was 99.40 ± 0.11% with a specific activity of 2.68 MBq/μg. [99mTc]Tc-HYNIC-KN035 displayed a high PD-L1 specificity both in vitro and in vivo and showed a high specific affinity for PD-L1 with an equilibrium dissociation constant (KD) of 31.04 nM. The binding of [99mTc]Tc-HYNIC-KN035 to H1975 cells (high expression of PD-L1) was much higher than to A549 cells (low expression of PD-L1). SPECT/CT imaging showed that H1975 tumors were visualized at 4 h post-injection and became clearer with time. However, mild tumor uptake was observed in A549 tumors and H1975 tumors of the blocking group at all time points. The uptake value of [99mTc]Tc-HYNIC-KN035 in H1975 tumors was increased continuously from 9.68 ± 0.91% ID/g at 4 h to 13.31 ± 2.23% ID/g at 24 h post-injection, which was higher than in A549 tumors with %ID/g of 4.59 ± 0.76 and 5.54 ± 0.28 at 4 and 24 h post-injection, respectively. These specific bindings were confirmed by blocking studies. [99mTc]Tc-HYNIC-KN035 can be synthesized easily and specifically targeted to PD-L1 in the tumor environment, allowing PD-L1 expression assessment noninvasively and dynamically with SPECT/CT imaging.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Nuclear Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Ying Ding
- Department of Nuclear Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.,Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.,Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan 430022, China
| | - Ning Li
- Department of Nuclear Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Sen Wang
- Department of Nuclear Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Si Zhou
- Department of Nuclear Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Ruping Li
- Department of Nuclear Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Hui Yang
- Department of Nuclear Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Wenliang Li
- Department of Nuclear Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Jinrong Qu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| |
Collapse
|
44
|
Sun Q, Li J, Ding Z, Liu Z. Radiopharmaceuticals heat anti-tumor immunity. Theranostics 2023; 13:767-786. [PMID: 36632233 PMCID: PMC9830438 DOI: 10.7150/thno.79806] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 01/06/2023] Open
Abstract
Radiopharmaceutical therapy (RPT) has proven to be an effective cancer treatment with minimal toxicity. With several RPT agents approved by FDA, the remarkable potential of this therapy is now being recognized, and the anti-tumor immunity induced by RPT is beginning to be noticed. This review evaluates the potential of RPT for immune activation, including promoting the release of danger associated-molecular pattern molecules that recruit inflammatory cells into the tumor microenvironment, and activating antigen-presenting cells and cytotoxic T cells. We also discuss the progress of combining RPT with immunotherapy to increase efficacy.
Collapse
Affiliation(s)
- Qi Sun
- Peking University-Tsinghua University Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jiyuan Li
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | | | - Zhibo Liu
- Peking University-Tsinghua University Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China,Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China,Changping Laboratory, Beijing 102206, China,✉ Corresponding author: Zhibo Liu:
| |
Collapse
|
45
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
46
|
Karizak AZ, Salmasi Z, Gheibihayat SM, Asadi M, Ghasemi Y, Tajbakhsh A, Savardashtaki A. Understanding the regulation of "Don't Eat-Me" signals by inflammatory signaling pathways in the tumor microenvironment for more effective therapy. J Cancer Res Clin Oncol 2023; 149:511-529. [PMID: 36342520 DOI: 10.1007/s00432-022-04452-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/22/2022] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Receptor/ligand pair immune checkpoints are inhibitors that regulate immunity as vital "Don't Find-Me" signals to the adaptive immune system, additionally, the essential goals of anti-cancer therapy. Moreover, the immune checkpoints are involved in treatment resistance in cancer therapy. The immune checkpoints as a signal from "self" and their expression on healthy cells prevent phagocytosis. Cells (e.g., senescent and/or apoptotic cells) with low immune checkpoints, such as low CD47 and/or PD-L1, are phagocytosed, which is necessary for tissue integrity and homeostasis maintenance. In other words, cancer cells induce increased CD47 expression in the tumor microenvironment (TME), avoiding their clearance by immune cells. PD-L1 and/or CD47 expression tumors have also been employed as biomarkers to guide cure prospects. Thus, targeting innate and adaptive immune checkpoints might improve the influence of the treatments on tumor cells. However, the CD47 regulation in the TME stands intricate, so much of this process has stayed a riddle. In this line, less attention has been paid to cytokines in TME. Cytokines are significant regulators of tumor immune surveillance, and they do this by controlling the actions of the immune cell. Recently, it has been suggested that different types of cytokines at TME might cooperate with others that contribute to the regulation of CD47 and/or PD-L1. MATERIALS AND METHODS The data were searched in available databases and a Web Search engine (PubMed, Scopus, and Google Scholar) using related keywords in the title, abstract, and keywords. CONCLUSION Given the significant role of pro/anti-inflammatory signaling in the TME, we discuss the present understanding of pro/anti-inflammatory signaling implications in "Don't Eat-Me" regulation signals, particularly CD47, in the pathophysiology of cancers and come up with innovative opinions for the clinical transformation and personalized medicine.
Collapse
Affiliation(s)
- Ashkan Zare Karizak
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Marzieh Asadi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 71362 81407, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran. .,Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 71362 81407, Iran. .,Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
47
|
Hasan N, Hasani NAH, Omar E, Sham FR, Fuad SBSA, Karim MKA, Ibahim MJ. A single targeted gamma-ray irradiation induced an acute modulation of immune cells and related cytokines in EMT6 mouse-bearing tumour model. Cancer Biomark 2023; 38:61-75. [PMID: 37522193 DOI: 10.3233/cbm-220268] [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: 08/01/2023]
Abstract
BACKGROUND A complicated interplay between radiation doses, tumour microenvironment (TME), and host immune system is linked to the active participation of immune response. OBJECTIVE The effects of single targeted 2 Gy and 8 Gy gamma-ray irradiations on the immune cell population (lymphocytes, B-cells, T-cells, neutrophils, eosinophils, and macrophages) in EMT6 mouse-bearing tumour models was investigated. METHODS The effects of both irradiation doses in early (96 hours) and acute phase (5 to 11 days) post-irradiation on immune parameters were monitored in blood circulation and TME using flow cytometry. Simultaneously, selected cytokines related to immune cells within the TME were measured using multiplex ELISA. RESULTS A temporary reduction in systemic total white blood count (TWBC) resulted from an early phase (96 hours) of gamma-ray irradiation at 2 Gy and 8 Gy compared to sham control group. No difference was obtained in the acute phase. Neutrophils dominated among other immune cells in TME in sham control group. Eosinophils in TME was significantly increased after 8 Gy treatment in acute phase compared to sham control (p< 0.005). Furthermore, the increment of tumour necrosis (TNF)-α, eotaxin and interleukin (IL)-7 (p< 0.05) in both treatment groups and phases were associated with anti-tumour activities within TME by gamma-ray irradiation. CONCLUSION The temporary changes in immune cell populations within systemic circulation and TME induced by different doses of gamma-ray irradiation correlated with suppression of several pro-tumorigenic cytokines in mouse-bearing EMT6 tumour models.
Collapse
Affiliation(s)
- Nurhaslina Hasan
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
- Faculty of Dentistry, University Teknologi MARA, Sungai Buloh, Selangor, Malaysia
| | | | - Effat Omar
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
| | - Fatihah Ronny Sham
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
| | | | | | | |
Collapse
|
48
|
The Lymphatic Endothelium in the Context of Radioimmuno-Oncology. Cancers (Basel) 2022; 15:cancers15010021. [PMID: 36612017 PMCID: PMC9817924 DOI: 10.3390/cancers15010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The study of lymphatic tumor vasculature has been gaining interest in the context of cancer immunotherapy. These vessels constitute conduits for immune cells' transit toward the lymph nodes, and they endow tumors with routes to metastasize to the lymph nodes and, from them, toward distant sites. In addition, this vasculature participates in the modulation of the immune response directly through the interaction with tumor-infiltrating leukocytes and indirectly through the secretion of cytokines and chemokines that attract leukocytes and tumor cells. Radiotherapy constitutes the therapeutic option for more than 50% of solid tumors. Besides impacting transformed cells, RT affects stromal cells such as endothelial and immune cells. Mature lymphatic endothelial cells are resistant to RT, but we do not know to what extent RT may affect tumor-aberrant lymphatics. RT compromises lymphatic integrity and functionality, and it is a risk factor to the onset of lymphedema, a condition characterized by deficient lymphatic drainage and compromised tissue homeostasis. This review aims to provide evidence of RT's effects on tumor vessels, particularly on lymphatic endothelial cell physiology and immune properties. We will also explore the therapeutic options available so far to modulate signaling through lymphatic endothelial cell receptors and their repercussions on tumor immune cells in the context of cancer. There is a need for careful consideration of the RT dosage to come to terms with the participation of the lymphatic vasculature in anti-tumor response. Here, we provide new approaches to enhance the contribution of the lymphatic endothelium to radioimmuno-oncology.
Collapse
|
49
|
Wang K, Yu HM, Xiang YJ, Cheng YQ, Ni QZ, Guo WX, Shi J, Feng S, Zhai J, Cheng SQ. Efficacy and safety of radiotherapy combined with atezolizumab plus bevacizumab in treating hepatocellular carcinoma with portal vein tumour thrombus: a study protocol. BMJ Open 2022; 12:e064688. [PMID: 36521893 PMCID: PMC9756192 DOI: 10.1136/bmjopen-2022-064688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/27/2022] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Vascular invasion and metastasis are poor prognostic factors in patients with hepatocellular carcinoma (HCC). The efficacy of available therapeutic regimens for unresectable HCC is not satisfactory in HCC with portal vein tumour thrombosis (PVTT). Therefore, this open-label, single-arm phase II clinical trial aims to investigate the efficacy and safety of radiotherapy combined with atezolizumab plus bevacizumab in treating HCC patients with PVTT. METHODS AND ANALYSIS We plan to enrol patients diagnosed with unresectable HCC complicated by PVTT. Intensity-modulated radiotherapy (IMRT) combined with atezolizumab plus bevacizumab will be administered for treatment. Patients will initially receive radiotherapy, with each IMRT cycle lasting for 28 days and the total dose of tumour (DT) of 40 Gy/20 f/26 d. CT scan will be performed again, and the treatment plan will be reformulated after field constriction. The treatment will continue until the total DT is up to 54-56 Gy/27-28 f. The treatment with atezolizumab plus bevacizumab will be started at 3±1 days after the initiation of radiotherapy and will continue until unacceptable toxicity or disease progression. The primary endpoint is objective response rate (ORR), while the secondary endpoints include overall survival, disease control rate, progression-free survival, time to progression, duration of response and the rate of surgical conversions. Assuming an ORR of 47%, with a two-sided alpha error of 0.1, 90% power, and a 10% drop-out rate, the required number of evaluable patients is 42. ETHICS AND DISSEMINATION This study will be conducted according to the standards of Good Clinical Practice and in compliance with the principles of the Declaration of Helsinki. The Ethics Committee of our Hospital has approved the protocol (EHBHKY2021-K-017). All participants are required to provide written informed consent. The results of the trial will be published in peer-reviewed journals and presented at international conferences. TRIAL REGISTRATION NUMBER ChiCTR2100049831.
Collapse
Affiliation(s)
- Kang Wang
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Hong-Ming Yu
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Yan-Jun Xiang
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Yu-Qiang Cheng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Qian-Zhi Ni
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Shanghai, China
| | - Wei-Xing Guo
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Jie Shi
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Shuang Feng
- Department of Radiotherapy, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Jian Zhai
- Department of Radiotherapy, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Shu-Qun Cheng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| |
Collapse
|
50
|
Zhu S, Wang Y, Tang J, Cao M. Radiotherapy induced immunogenic cell death by remodeling tumor immune microenvironment. Front Immunol 2022; 13:1074477. [PMID: 36532071 PMCID: PMC9753984 DOI: 10.3389/fimmu.2022.1074477] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
Emerging evidence indicates that the induction of radiotherapy(RT) on the immunogenic cell death (ICD) is not only dependent on its direct cytotoxic effect, changes in the tumor immune microenvironment also play an important role in it. Tumor immune microenvironment (TIME) refers to the immune microenvironment that tumor cells exist, including tumor cells, inflammatory cells, immune cells, various signaling molecules and extracellular matrix. TIME has a barrier effect on the anti-tumor function of immune cells, which can inhibit all stages of anti-tumor immune response. The remodeling of TIME caused by RT may affect the degree of immunogenicity, and make it change from immunosuppressive phenotype to immunostimulatory phenotype. It is of great significance to reveal the causes of immune escape of tumor cells, especially for the treatment of drug-resistant tumor. In this review, we focus on the effect of RT on the TIME, the mechanism of RT in reversing the TIME to suppress intrinsic immunity, and the sensitization effect of the remodeling of TIME caused by RT on the effectiveness of immunotherapy.
Collapse
|