1
|
Chen Y, Zhou C, Zhang X, Chen M, Wang M, Zhang L, Chen Y, Huang L, Sun J, Wang D, Chen Y. Construction of a novel radioresistance-related signature for prediction of prognosis, immune microenvironment and anti-tumour drug sensitivity in non-small cell lung cancer. Ann Med 2025; 57:2447930. [PMID: 39797413 PMCID: PMC11727174 DOI: 10.1080/07853890.2024.2447930] [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: 06/30/2024] [Revised: 11/26/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a fatal disease, and radioresistance is an important factor leading to treatment failure and disease progression. The objective of this research was to detect radioresistance-related genes (RRRGs) with prognostic value in NSCLC. METHODS The weighted gene coexpression network analysis (WGCNA) and differentially expressed genes (DEGs) analysis were performed to identify RRRGs using expression profiles from TCGA and GEO databases. The least absolute shrinkage and selection operator (LASSO) regression and random survival forest (RSF) were used to screen for prognostically relevant RRRGs. Multivariate Cox regression was used to construct a risk score model. Then, Immune landscape and drug sensitivity were evaluated. The biological functions exerted by the key gene LBH were verified by in vitro experiments. RESULTS Ninety-nine RRRGs were screened by intersecting the results of DEGs and WGCNA, then 11 hub RRRGs associated with survival were identified using machine learning algorithms (LASSO and RSF). Subsequently, an eight-gene (APOBEC3B, DOCK4, IER5L, LBH, LY6K, RERG, RMDN2 and TSPAN2) risk score model was established and demonstrated to be an independent prognostic factor in NSCLC on the basis of Cox regression analysis. The immune landscape and sensitivity to anti-tumour drugs showed significant disparities between patients categorized into different risk score subgroups. In vitro experiments indicated that overexpression of LBH enhanced the radiosensitivity of A549 cells, and knockdown LBH reversed the cytotoxicity induced by X-rays. CONCLUSION Our study developed an eight-gene risk score model with potential clinical value that can be adopted for choice of drug treatment and prognostic prediction. Its clinical routine use may assist clinicians in selecting more rational practices for individuals, which is important for improving the prognosis of NSCLC patients. These findings also provide references for the development of potential therapeutic targets.
Collapse
Affiliation(s)
- Yanliang Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Chan Zhou
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaoqiao Zhang
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Min Chen
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Meifang Wang
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Lisha Zhang
- Department of Obstetrics, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Yanhui Chen
- Department of Neuroscience and Endocrinology, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Litao Huang
- Department of Clinical Research Management, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junjun Sun
- Department of Emergency Surgery, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, , China
| | - Dandan Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Yong Chen
- Department of Radio-Chemotherapy, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
2
|
Zhang A, Fan L, Liu Q, Zuo X, Zhu J. Immunological Effects of Proton Radiotherapy: New Opportunities and Challenges in Cancer Therapy. CANCER INNOVATION 2025; 4:e70003. [PMID: 40061827 PMCID: PMC11885950 DOI: 10.1002/cai2.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/23/2024] [Accepted: 01/16/2025] [Indexed: 03/14/2025]
Abstract
Radiation therapy can be categorised by particle type into photon, proton and heavy ion therapies. Proton radiotherapy is highlighted due to its unique physical properties, such as the Bragg peak and minimal exit dose, which offer superior dose distribution. This makes proton radiotherapy especially advantageous for treating tumours near vital organs with complex structures, such as gliomas near the brain, nasopharyngeal carcinoma near the brainstem and mediastinal tumours near the heart. Proton irradiation can induce distant effects through immunogenicity within the target area. The reduced low-dose zone outside the target provides better lymphatic system protection and immune benefits. Additionally, combining proton radiotherapy with immunotherapy may offer further biological advantages. These features make proton radiotherapy a promising option in cancer treatment. This article may aid in the understanding of proton radiotherapy and its immune effects and lead to new effective options for tumour treatment.
Collapse
Affiliation(s)
- Anhang Zhang
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| | - Liyuan Fan
- Department of Radiation OncologyQilu Hospital of Shandong UniversityJinanShandongChina
| | - Qi Liu
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| | - Xiaoxin Zuo
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| | - Jian Zhu
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| |
Collapse
|
3
|
Lan J, Cai D, Gou S, Bai Y, Lei H, Li Y, Chen Y, Zhao Y, Shen J, Wu X, Li M, Chen M, Li X, Sun Y, Gu L, Li W, Wang F, Cho CH, Zhang Y, Zheng X, Xiao Z, Du F. The dynamic role of ferroptosis in cancer immunoediting: Implications for immunotherapy. Pharmacol Res 2025; 214:107674. [PMID: 40020885 DOI: 10.1016/j.phrs.2025.107674] [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: 12/19/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
Currently, cancer immunotherapy strategies are primarily formulated based on the patient's present condition, representing a "static" treatment approach. However, cancer progression is inherently "dynamic," as the immune environment is not fixed but undergoes continuous changes. This dynamism is characterized by the ongoing interactions between tumor cells and immune cells, which ultimately lead to alterations in the tumor immune microenvironment. This process can be effectively elucidated by the concept of cancer immunoediting, which divides tumor development into three phases: "elimination," "equilibrium," and "escape." Consequently, adjusting immunotherapy regimens based on these distinct phases may enhance patient survival and improve prognosis. Targeting ferroptosis is an emerging area in cancer immunotherapy, and our findings reveal that the antioxidant systems associated with ferroptosis possess dual roles, functioning differently across the three phases of cancer immunoediting. Therefore, this review delve into the dual role of the ferroptosis antioxidant system in tumor development and progression. It also propose immunotherapy strategies targeting ferroptosis at different stages, ultimately aiming to illuminate the significant implications of targeting ferroptosis at various phases for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiarui Lan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Dan Cai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Shuang Gou
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Yulin Bai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Huaqing Lei
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yan Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yan Zhang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China
| | - Xin Zheng
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China.
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| |
Collapse
|
4
|
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] [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
|
5
|
Deng S, Hu L, Chen G, Ye J, Xiao Z, Guan T, Guo S, Xia W, Cheng D, Wan X, Cheng K, Ou C. A PD-L1 siRNA-Loaded Boron Nanoparticle for Targeted Cancer Radiotherapy and Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2419418. [PMID: 39955653 DOI: 10.1002/adma.202419418] [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: 12/11/2024] [Revised: 01/25/2025] [Indexed: 02/17/2025]
Abstract
Although the combination of radiotherapy and immunotherapy is regarded as a promising clinical treatment strategy, numerous clinical trials have failed to demonstrate synergistic effects. One of the key reasons is that conventional radiotherapies inevitably damage intratumoral effector immune cells. Boron Neutron Capture Therapy (BNCT) is a precise radiotherapy that selectively kills tumor cells while sparing adjacent normal cells, by utilizing 10B agents and neutron irradiation. Therefore, combinational BNCT-immunotherapy holds promise for achieving more effective synergistic effects. Here it develops a 10B-containing polymer that self-assembled with PD-L1 siRNA to form 10B/siPD-L1 nanoparticles for combinational BNCT-immunotherapy. Unlike antibodies, PD-L1 siRNA can inhibit intracellular PD-L1 upregulated by BNCT, activating T-cell immunity while also suppressing DNA repair. This can enhance BNCT-induced DNA damage, promoting immunogenic cell death (ICD) and further amplifying the antitumor immune effect. The results demonstrated that BNCT using 10B/siPD-L1 nanoparticles precisely killed tumor cells while sparing adjacent T cells and induced a potent antitumor immune response, inhibiting distal and metastatic tumors.
Collapse
Affiliation(s)
- Shaohui Deng
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Lijun Hu
- The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Guo Chen
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jujian Ye
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zecong Xiao
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Tianwang Guan
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Shuai Guo
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Wei Xia
- Neuboron Medtech Ltd, Nanjing, 211112, China
| | - Du Cheng
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Xiaochun Wan
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York, 10032, USA
| | - Caiwen Ou
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| |
Collapse
|
6
|
Liu J, Gong Y, Wang D, Kang S, Gong S, Ma H, Gong P, Kong B. Water-Dispersible MXene Governs Glycolysis for Cancer Synergistic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2411768. [PMID: 40159863 DOI: 10.1002/smll.202411768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/24/2025] [Indexed: 04/02/2025]
Abstract
Targeted delivery of glucose oxidase (GOx) using MXene remains a great challenge due to its poor dispersion and susceptibility to oxidation, and the hypoxia and high glutathione (GSH) contents make the situation even more worrying. Herein, a bovine serum albumin-mediated non-chemical modification strategy is developed, endowing titanium carbide MXene with long-time water-dispersion and further integrating it as a glycolysis-controllable therapy system without any chemotherapeutic agents. The system also constructs an effective O2 cycling and GSH degradation pathway, which fundamentally adjusts the tumor microenvironment and greatly elevates both in vivo and in vitro therapy effects. Reactive oxygen species are also generated and disrupt the balance of oxidative stress. Moreover, the reduced efficiency of mitochondrial energy production significantly inhibits the level of glycolysis and hinders energy supply. The study presents an effective cancer treatment combining starvation/photothermal therapy, which has superior anti-cancer effects due to the dual effects of reducing glucose levels and diminishing cellular energy production capacity.
Collapse
Affiliation(s)
- Jinfeng Liu
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong, 273165, P. R. China
| | - Yuwen Gong
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong, 273165, P. R. China
| | - Dandan Wang
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong, 273165, P. R. China
| | - Shuangli Kang
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong, 273165, P. R. China
| | - Shengjian Gong
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong, 273165, P. R. China
| | - Hanqing Ma
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong, 273165, P. R. China
| | - Peiwei Gong
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong, 273165, P. R. China
| | - Biao Kong
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| |
Collapse
|
7
|
Lei L, Xu H, Li M, Du M, Chen Z. Dual-pathway tumor radiosensitization strategy based on engineered bacteria capable of targeted delivery of AuNPs and specific hypoxia alleviation. J Nanobiotechnology 2025; 23:254. [PMID: 40155884 PMCID: PMC11954313 DOI: 10.1186/s12951-025-03329-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/14/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Radiotherapy efficacy remains constrained by two key challenges: dose-dependent toxicity to healthy tissues at high radiation doses and hypoxia-mediated tumor radioresistance. While radiosensitizers like gold nanoparticles can enhance tumor-specific radiation deposition, their targeted delivery to tumors presents a significant hurdle. Bacteria have emerged as promising bio-carriers that not only actively target tumors and penetrate complex microenvironments, but can also be genetically engineered as multifunctional platforms for radiosensitizer delivery and hypoxia alleviation. RESULTS An integrated nanosystem (PCM@AuNPs), composed of engineered bacteria (PCM) and gold nanoparticles (AuNPs), is used to increase the effectiveness of radiotherapy. PCM can target and colonize tumor sites more effectively, thus improving the delivery efficiency of radiosensitizers. Furthermore, PCM overexpresses catalase (CAT), which decomposes excess H2O2 into O2, helping to mitigate hypoxia in the TME. Under X-ray irradiation, PCM@AuNPs significantly enhance radiosensitization, leading to improved tumor growth inhibition while maintaining good biocompatibility. CONCLUSIONS An effective strategy based on an integrated nanosystem (PCM@AuNPs) for radiosensitization through multiple pathways is developed. This novel engineered bacterial strategy holds great promise for enhancing radiosensitization in cancer therapy.
Collapse
Affiliation(s)
- Lingling Lei
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, Changsha, China
- Department of Medical Imaging, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, Changsha, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| | - Haonan Xu
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, Changsha, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- School of Public Health, University of South China, Hengyang, China
| | - Mingjie Li
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, Changsha, China
- Department of Medical Imaging, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, Changsha, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| | - Meng Du
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, Changsha, China.
- Department of Medical Imaging, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, Changsha, China.
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China.
| | - Zhiyi Chen
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, Changsha, China.
- Department of Medical Imaging, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, Changsha, China.
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
8
|
Rahimi A, Baghernejadan Z, Hazrati A, Malekpour K, Samimi LN, Najafi A, Falak R, Khorramdelazad H. Combination therapy with immune checkpoint inhibitors in colorectal cancer: Challenges, resistance mechanisms, and the role of microbiota. Biomed Pharmacother 2025; 186:118014. [PMID: 40157004 DOI: 10.1016/j.biopha.2025.118014] [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: 01/08/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Colorectal cancer (CRC) is still one of the leading causes of cancer deaths worldwide. Even though there has been progress in cancer immunotherapy, the results of applying immune checkpoint inhibitors (ICIs) have been unsatisfactory, especially in microsatellite stable (MSS) CRC. Single-agent ICIs that target programmed cell death-1 (PD-1)/ PD-L1, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), T cell Ig- and mucin-domain-containing molecule-3 (TIM-3), and lymphocyte activation gene (LAG)-3 have emerged as having specific benefits. However, many primary and secondary resistance mechanisms are available in the tumor microenvironment (TME) that prevent it from happening. Combination strategies, such as the use of anti-PD-1 and anti-CTLA-4, can be effective in overcoming these resistance pathways, but toxicities remain a significant concern. Moreover, ICIs have been integrated with various treatment modalities, including chemotherapy, radiotherapy, antibiotics, virotherapy, polyadenosine diphosphate-ribose polymerase (PARP) inhibitors, and heat shock protein 90 (HSP90) inhibitors. The outcomes observed in both preclinical and clinical settings have been encouraging. Interestingly, manipulating gut microbiota via fecal microbiota transplantation (FMT) has been identified as a new strategy to increase the efficacy of immunotherapy in CRC patients. Therefore, integrating ICIs with other treatment approaches holds promise in enhancing the prognosis of CRC patients. This review focuses on the unmet need for new biomarkers to select patients for combination therapies and the ongoing work to overcome resistance and immune checkpoint blockade.
Collapse
Affiliation(s)
- Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Baghernejadan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alireza Najafi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
9
|
Ruder S, Ricaurte-Fajardo A, Sun M, Castellanos SH, Osborne JR, Tagawa ST. Advances in PSMA-Targeted Radionuclide Therapeutics. Curr Treat Options Oncol 2025:10.1007/s11864-025-01296-7. [PMID: 40138150 DOI: 10.1007/s11864-025-01296-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2025] [Indexed: 03/29/2025]
Abstract
OPINION STATEMENT Prostate-specific membrane antigen targeted radionuclide therapies (PSMA-TRT) such as 177Lu-PSMA-617 hold great promise in improving clinical outcomes at various stages of prostate cancer. The FDA approval of 177Lu-PSMA-617 represents a significant advancement in the treatment of metastatic castration-resistant prostate cancer (mCRPC). The VISION trial demonstrated improved radiographic progression-free survival (rPFS) and overall survival (OS) with 177Lu-PSMA-617 in patients with mCRPC who had already receive androgen receptor pathway inhibitor (ARPI) and taxane chemotherapy. Exploration of 177Lu-PSMA-617 in earlier stages of prostate cancer, such as in the PSMAfore trial for patients who have not received chemotherapy, holds great promise for improving long-term outcomes and delaying exposure to chemotherapy. Combining 177Lu-PSMA-617 with other therapies, including chemotherapy, PARP inhibitors, and immunotherapy, is an area of active investigation. This review will also discuss alternative radionuclides (such as actininum-225 and terbium-161) and delivery vehicles (such as PSMA-I&T), which we find promising. Predictive biomarkers and dosimetry will be crucial for identifying patients most likely to benefit from PSMA-TRT. Continued research and refinement of these therapies will lead to PSMA-targeted treatments becoming an integral part of prostate cancer management.
Collapse
Affiliation(s)
- Samuel Ruder
- Department of Medicine, Division of Hematology and Oncology, New York Presbyterian Weill Cornell Medical Center, 520 East 70th Street, Starr Pavilion, NY, NY, 10065, USA.
| | - Andres Ricaurte-Fajardo
- Department of Radiology, Division of Molecular Imaging and Therapeutics, Weill Cornell Medicine, New York, NY, USA
| | - Michael Sun
- Department of Medicine, Division of Hematology and Oncology, New York Presbyterian Weill Cornell Medical Center, 520 East 70th Street, Starr Pavilion, NY, NY, 10065, USA
| | - Sandra Huicochea Castellanos
- Department of Radiology, Division of Molecular Imaging and Therapeutics, Weill Cornell Medicine, New York, NY, USA
| | - Joseph R Osborne
- Department of Radiology, Division of Molecular Imaging and Therapeutics, Weill Cornell Medicine, New York, NY, USA
| | - Scott T Tagawa
- Department of Medicine, Division of Hematology and Oncology, New York Presbyterian Weill Cornell Medical Center, 520 East 70th Street, Starr Pavilion, NY, NY, 10065, USA.
| |
Collapse
|
10
|
Geboers B, Timmer F, Vos D, Scheffer H, Bakker J, Ruarus A, Vroomen L, Stam A, Lougheed S, Schouten E, Puijk R, van den Tol P, Lagerwaard F, de Vries J, Bruynzeel A, Meijerink M, de Gruijl T. Systemic immunomodulation by irreversible electroporation versus stereotactic ablative body radiotherapy in locally advanced pancreatic cancer: the CROSSFIRE trial. J Immunother Cancer 2025; 13:e010222. [PMID: 40139834 PMCID: PMC11950998 DOI: 10.1136/jitc-2024-010222] [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: 07/31/2024] [Accepted: 01/28/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Irreversible electroporation (IRE) and stereotactic ablative body radiotherapy (SABR) are cytoreductive therapies for locally advanced pancreatic cancer (LAPC). Both may signify immunogenic cell death. We aimed to compare systemic immune responses between the treatments. METHODS As part of the randomized phase II CROSSFIRE trial (NCT02791503), comparing the oncological efficacy of IRE to SABR in patients with LAPC, pre- and post-treatment (2 weeks and 3 months) peripheral blood samples were collected. Frequency and activation status of lymphocytic and myeloid subsets were determined using flow cytometry. T cell responses to pancreatic cancer associated with Wilms tumor-1 (WT-1) and survivin tumor antigens were determined by interferon-γ enzyme-linked immunospot assay. RESULTS In total, 20 IRE and 20 SABR-treated participants were analyzed (20 men; median age 65 (IQR 55-70)). IRE induced immediate decreases in systemic regulatory T cell (Treg) and conventional type-1 dendritic cell rates, coinciding with CD4+/CD8+ T cell activation by upregulation of PD-1, which was associated with improved overall survival (OS). SABR similarly induced immediate CD4+/CD8+ T cell activation by upregulation of Ki67 and CD25 but resulted in asynchronously delayed Treg downregulation. SABR also induced a durable increase in CD4+ EM T cells, associated with improved OS. Ablation-induced WT-1 or survivin-specific T cell responses were observed in 9/16 (56%) immune competent participants (IRE n=5, SABR n=4) and were associated with longer OS. CONCLUSION Distinct immune stimulatory responses associated with improved OS, suggest that SABR might benefit from combined Treg depletion strategies while IRE could benefit from PD-1 checkpoint inhibition. TRIAL REGISTRATION NUMBER The trial was registered on clinical trials.gov (NCT02791503).
Collapse
Affiliation(s)
- Bart Geboers
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Medical Imaging, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Florentine Timmer
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Danielle Vos
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Hester Scheffer
- Department of Radiology, Noord West Ziekenhuis Groep, Alkmaar, The Netherlands
| | - Joyce Bakker
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
| | - Alette Ruarus
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Laurien Vroomen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | | | | | - Evelien Schouten
- Department of Radiotherapy, Antoni van Leeuwenhoek Hospital - Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Robbert Puijk
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, OLVG, Amsterdam, The Netherlands
| | | | - Frank Lagerwaard
- Department of Radiation Oncology, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
| | - Jan de Vries
- Department of Radiology and Nuclear Medicine, OLVG, Amsterdam, The Netherlands
| | - Anna Bruynzeel
- Department of Radiation Oncology, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
| | - Martijn Meijerink
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Tanja de Gruijl
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
| |
Collapse
|
11
|
Wu G, Liu Y, Fan H, Rao M, Zhang J, Zhang J. Tislelizumab plus anlotinib with or without radiotherapy as first-line therapy in advanced hepatocellular carcinoma: a single center, non-randomized retrospective case-control study. Discov Oncol 2025; 16:387. [PMID: 40131659 PMCID: PMC11937455 DOI: 10.1007/s12672-025-02171-5] [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: 09/18/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
The purpose of this study was to investigate the efficacy and safety of tislelizumab (monoclonal antibody) plus anlotinib (tyrosine kinase inhibitor) with or without radiotherapy in advanced hepatocellular carcinoma (HCC). Ninety patients with advanced HCC were divided into two groups: tislelizumab plus anlotinib with radiotherapy (TAR group) and tislelizumab plus anlotinib (TA group) based on the treatment received. Radiotherapy was performed on two or three days during the first cycle of tislelizumab plus anlotinib. The radiotherapy requirements were dose95% ≥ 14.2-46 Gy for tumor volume. Efficacy was evaluated according to the modified Response Evaluation Criteria for Solid Tumors. Adverse events (AEs) were evaluated using the National Cancer Institute-Common Terminology Criteria for Adverse Events 4.0. The primary endpoint was the objective response rate (ORR). The secondary endpoints were progression-free survival (PFS), overall survival (OS), and the disease control rate (DCR). The ORR and DCR in the TAR group were 24.5% (62.2% and 37.7%, p = 0.03) and 22.3% higher (75.6% and 53.3%, p = 0.04), respectively, compared to the TA group. The median OS and PFS in the TAR group were prolonged 4.5 months [21.0 and 16.5 months, χ2 = 8.99, p = 0.00, 95% confidence interval (CI) 0.295-0.774] and 4.0 months (11.0 and 7.0 months. χ2 = 11.73, p = 0.00. 95% CI 0.989-2.502), respectively, compared to the TA group. The risks of disease progression and mortality in the TAR group were reduced by 53.0% (hazard ratio (HR) = 0.470, 95% CI 0.294-0.753) and 49.3% (HR = 0.507, 95% CI 0.315-0.815) compared to the TA group. The OS and PFS rates at 1 and 2 years increased by 28.9% (97.8% and 68.9%, p = 0.00) and 20.0% (42.2% and 22.2%, p = 0.07) and 28.9% (42.2% and 13.3%, p = 0.00) and 15.6% (20.0% and 4.4%, p = 0.05), respectively, in the TAR group compared to the TA group. Most patients mainly presented with grade 1/2 tolerable acute AEs (p > 0.05). No AEs were related to radiotherapy, and no fatalities occurred. The results indicate that tislelizumab plus anlotinib and radiotherapy is a safe and effective treatment for advanced HCC. Trial registration: ChiCTR2000039022 (10/13/2020). Retrospective.
Collapse
Affiliation(s)
- Guishu Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yuhong Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Huaxi Fan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Mingyue Rao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jing Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jianwen Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, People's Republic of China.
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
12
|
Burckel H, Nicol A, Mura C, Rousseau M, Bou-Gharios J, Froidurot L, Richard C, Morgand V, Laurent PA, Limagne E, Boidot R, Noël G, Mirjolet C. Distinct immune responses to proton and photon radiotherapy: implications for anti-PD-L1 combination therapy in colorectal cancer. J Transl Med 2025; 23:360. [PMID: 40122794 PMCID: PMC11931879 DOI: 10.1186/s12967-025-06377-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Ionizing radiation can influence the antitumor immune response, either activating or suppressing the immune system depending on the tumor type and radiotherapy modality. While photon radiation (RT) combined with immunotherapy (IT) is widely studied in clinical trials, proton radiation (PT) combined with IT has not been thoroughly investigated in clinical or preclinical studies despite its radiobiological advantages. This study aims to explore the immune effects of a hypofractionated PT scheme compared to RT and its efficacy with anti-PD-L1 immunotherapy. METHODS Balb/c mice bearing subcutaneous CT26 colon tumors were treated with RT or PT, delivered with 3 × 8 Gy. Seven days post-treatment, transcriptomic analysis and immune response assessments to characterize lymphoid cells, myeloid cells, and PD-L1 expression were performed. Tumor growth was monitored to evaluate the efficacy of combining RT or PT with anti-PD-L1 IT. RESULTS The RNA sequencing analysis demonstrated an overexpression of genes involved in the interferon type I pathway after both RT and PT. Tumor microenvironment analysis showed enhanced immune cell infiltration in tumors after both treatments. Immunoactivating cells infiltration was observed, with LT CD8 + cells infiltration after both RT and PT, more significantly after RT. NK and TAM1 cells infiltrated only after RT. Immunosuppressive cell populations were induced by PT, including MDSCs, while Tregs infiltrated both RT and PT treated tumors. PD-L1 expression was significantly induced only by RT. The combination of anti-PD-L1 with RT or PT resulted in tumor growth delay compared to RT or PT alone, with a significant survival benefit observed only after the combination of RT and IT. CONCLUSIONS This study demonstrates that hypofractionated RT and PT induced both similar and significantly distinct immune responses. PT triggers a stronger immunosuppressive response than RT. Optimizing the combination of PT with IT, including dose, fractionation, and sequencing is crucial for improving treatment efficacy.
Collapse
Affiliation(s)
- Hélène Burckel
- Radiobiology Laboratory, Paul Strauss Comprehensive, Cancer Center, Institut de Cancérologie Strasbourg Europe (ICANS), UNICANCER, 67000, Strasbourg, France.
- Equipe Imagerie Multimodale Intégrative en Santé, ICube, UMR7357, Université de Strasbourg, Strasbourg, France.
| | - Anaïs Nicol
- Radiobiology Laboratory, Paul Strauss Comprehensive, Cancer Center, Institut de Cancérologie Strasbourg Europe (ICANS), UNICANCER, 67000, Strasbourg, France
- Equipe Imagerie Multimodale Intégrative en Santé, ICube, UMR7357, Université de Strasbourg, Strasbourg, France
| | - Carole Mura
- Radiobiology Laboratory, Paul Strauss Comprehensive, Cancer Center, Institut de Cancérologie Strasbourg Europe (ICANS), UNICANCER, 67000, Strasbourg, France
- Equipe Imagerie Multimodale Intégrative en Santé, ICube, UMR7357, Université de Strasbourg, Strasbourg, France
| | - Marc Rousseau
- Institut Pluridisciplinaire Hubert Curien, Université de Strasbourg, CNRS, UMR 7178, 67200, Strasbourg, France
| | - Jolie Bou-Gharios
- Radiobiology Laboratory, Paul Strauss Comprehensive, Cancer Center, Institut de Cancérologie Strasbourg Europe (ICANS), UNICANCER, 67000, Strasbourg, France
- Equipe Imagerie Multimodale Intégrative en Santé, ICube, UMR7357, Université de Strasbourg, Strasbourg, France
| | - Lisa Froidurot
- X-Rain: Research Unit in Radiotherapy Combined with Immunotherapies and Nanoparticles, IMATHERA, Department of Radiation Oncology, Unicancer-Georges-Francois Leclerc Cancer Center, 21000, Dijon, France
| | - Corentin Richard
- Molecular Biology Clinical Research, Unicancer-Center Georges-Francois Leclerc, 21000, Dijon, France
| | - Véronique Morgand
- X-Rain: Research Unit in Radiotherapy Combined with Immunotherapies and Nanoparticles, IMATHERA, Department of Radiation Oncology, Unicancer-Georges-Francois Leclerc Cancer Center, 21000, Dijon, France
| | - Pierre-Antoine Laurent
- X-Rain: Research Unit in Radiotherapy Combined with Immunotherapies and Nanoparticles, IMATHERA, Department of Radiation Oncology, Unicancer-Georges-Francois Leclerc Cancer Center, 21000, Dijon, France
| | - Emeric Limagne
- CTM, INSERM, UMR 1231, Trecs Team, CTM, 21000, Dijon, France
- PTBC, CGFL, Dijon, France
| | - Romain Boidot
- Molecular Biology Clinical Research, Unicancer-Center Georges-Francois Leclerc, 21000, Dijon, France
| | - Georges Noël
- Radiobiology Laboratory, Paul Strauss Comprehensive, Cancer Center, Institut de Cancérologie Strasbourg Europe (ICANS), UNICANCER, 67000, Strasbourg, France
- Equipe Imagerie Multimodale Intégrative en Santé, ICube, UMR7357, Université de Strasbourg, Strasbourg, France
- Institut de Cancérologie Strasbourg Europe (ICANS), Paul Strauss Comprehensive Cancer Center, Department of Radiation Oncology, UNICANCER, 17 Rue Albert Calmette, 67200, Strasbourg, France
| | - Céline Mirjolet
- X-Rain: Research Unit in Radiotherapy Combined with Immunotherapies and Nanoparticles, IMATHERA, Department of Radiation Oncology, Unicancer-Georges-Francois Leclerc Cancer Center, 21000, Dijon, France
- CTM, INSERM, UMR 1231, Trecs Team, CTM, 21000, Dijon, France
| |
Collapse
|
13
|
Zhang YP, Guo ZQ, Cai XT, Rong ZX, Fang Y, Chen JQ, Zhuang KM, Ruan MJ, Ma SC, Lin LY, Han DD, Li YS, Wang YY, Wang J, Cao CH, Tang XR, Xie QK, Chen Y, Lin Y, Tan JL, Yu ZH, Wu ZN, Wei W, Zheng DY, Zeng YJ, Ruan YC, Xu ZP, Gu JZ, Xiao LS, Liu L, Guan J, Bai X, Wu DH, Dong ZY. PAI-1-driven SFRP2 high cancer-associated fibroblasts hijack the abscopal effect of radioimmunotherapy. Cancer Cell 2025:S1535-6108(25)00076-5. [PMID: 40086438 DOI: 10.1016/j.ccell.2025.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/15/2024] [Accepted: 02/20/2025] [Indexed: 03/16/2025]
Abstract
The abscopal effect of radioimmunotherapy, wherein tumor shrinkage occurs beyond the irradiated field, is therapeutically promising but clinically rare. The mechanisms underlying this effect remain elusive. Here, in vivo genome-wide CRISPR screening identifies SFRP2 as a potential stromal regulator of the abscopal effect. SFRP2 exhibits cancer-associated fibroblast (CAF)-specific expression and radioimmunotherapy-mediated upregulation in unirradiated tumors. Conditional Sfrp2 knockout in CAFs boosts the abscopal effect by rewiring the vascular-immune microenvironment to promote CD8+ T cell recruitment to unirradiated tumors. In vivo lineage tracing reveals that elevated SFRP2 correlates with radioimmunotherapy-driven pericyte lineage commitment. Serum proteomics reveals that irradiated-tumor-secreted PAI-1 triggers distant tumor pericyte cell-fate transition into SFRP2high CAFs via the LRP1/p65 axis. Pharmacologically blocking SFRP2 or PAI-1 enhances the abscopal effect in humanized patient-derived xenograft models. Our findings collectively illustrate that PAI-1-induced SFRP2high CAFs serve as critical stromal regulator to hijack the abscopal effect, providing promising targets for enhancing radioimmunotherapy effectiveness.
Collapse
Affiliation(s)
- Yan-Pei Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ze-Qin Guo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Ting Cai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zi-Xuan Rong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuan Fang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jia-Qi Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kui-Mao Zhuang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Min-Jie Ruan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Si-Cong Ma
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Le-Yi Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Duan-Duan Han
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yang-Si Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yuan-Yuan Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jian Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chuan-Hui Cao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xin-Ran Tang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qian-Kun Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yue Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yan Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jia-Le Tan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zi-Hang Yu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ze-Nan Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wei Wei
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, Hubei 441000, China
| | - Da-Yong Zheng
- Department of Oncology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Shunde 528333, China
| | - Yu-Jie Zeng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Ying-Chen Ruan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zi-Peng Xu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jun-Zi Gu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lu-Shan Xiao
- Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Li Liu
- Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Health Management Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jian Guan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Xue Bai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - De-Hua Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Zhong-Yi Dong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
14
|
Zhang S, Wang C, Zhu Y, Gao J, Yan Y, Chen M, Yan X, Liu Z, Feng L. DNA-Capturing Manganese-Coordinated Chitosan Microparticles Potentiate Radiotherapy via Activating the cGAS-STING Pathway and Maintaining Tumor-Infiltrating CD8 + T-Cell Stemness. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2418583. [PMID: 39955699 DOI: 10.1002/adma.202418583] [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: 11/28/2024] [Revised: 02/03/2025] [Indexed: 02/17/2025]
Abstract
The radiotherapy-induced release of DNA fragments can stimulate the cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) pathway to prime antitumor immunity, but this pathway is expected to be less potent because of the inefficient cytosolic delivery of negatively charged DNA fragments. In this study, manganese-coordinated chitosan (CS-Mn) microparticles with selective DNA-capturing capacity are concisely prepared via a coordination-directed one-pot synthesis process to potentiate the immunogenicity of radiotherapy. The obtained CS-Mn microparticles that undergo rapid disassembly under physiological conditions can selectively bind with DNA to form positively charged DNA-CS assemblies because of the strong electrostatic interaction between linear chitosan and DNA molecules. They thus enable efficient cytosolic delivery of DNA in the presence of serum to cooperate with Mn2+ to activate the cGAS-STING pathway in dendritic cells. Upon intratumoral injection, the CS-Mn microparticles markedly enhance the efficacy of radiotherapy against both irradiated and distal tumors in different tumor models via collectively promoting tumor-infiltrating CD8+ T-cell stemness and the activation of innate immunity. The radiosensitization effect of CS-Mn microparticles can be further augmented by concurrently applying anti-programmed cell death protein 1 (anti-PD-1) immunotherapy. This work highlights an ingenious strategy to prepare Trojan horse-like DNA-capturing microparticles as cGAS-STING-activating radiosensitizers for effective radioimmunotherapy.
Collapse
Affiliation(s)
- Shuai Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Chunjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Yujie Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Juxin Gao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Yifan Yan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Minming Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Xiaoying Yan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Liangzhu Feng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| |
Collapse
|
15
|
Pham TN, Coupey J, Thariat J, Valable S. Impact of circulating lymphocyte kinetics following radiotherapy on patient survival: A model-based meta-analysis. Comput Biol Med 2025; 186:109702. [PMID: 39864332 DOI: 10.1016/j.compbiomed.2025.109702] [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: 11/05/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/28/2025]
Abstract
INTRODUCTION Radiation-induced lymphopenia (RIL) has been shown to adversely affect the prognosis of cancer patients undergoing radiotherapy (RT). This model-based meta-analysis investigated the prognostic significance of lymphocyte counts both early and late after RT and examined the dose‒response relationship between post-RT lymphocyte levels and patient survival. METHODS A literature search of published articles on the effect of RIL on cancer prognosis was conducted using the PubMed and Cochrane databases. A survival model was developed, incorporating absolute lymphocyte count (ALC) thresholds during (1 month after RT initiation, nadir) and 6 months after RT (recovery) as covariates to estimate progression-free survival (PFS) and overall survival (OS). This survival model, with the lymphocyte count cutoff as a covariate, was then used to simulate the benefit of increased PFS and OS in populations without lymphopenia or severe lymphopenia compared to the total population. RESULTS A total of 35 studies met the inclusion criteria for survival analysis. Our survival model revealed an increase in survival in the subgroup without lymphopenia compared to the total population. The subgroup without lymphopenia 1 month after RT initiation showed a 10.28 % and 3.92 % increase in 24-month PFS and OS, respectively. The subgroup without lymphopenia at 6 months showed a 5.82 % and 2.78 % increase in 24-month PFS and OS, respectively. CONCLUSION This study highlights the critical role of lymphocyte nadir and recovery following RT in patient prognosis and strengthens the evidence for a causal relationship between RIL and patient outcomes. Expanding the dataset and including randomized controlled trials would provide a more comprehensive understanding of monitoring or knowledge of ALC profiles following RT.
Collapse
Affiliation(s)
- Thao-Nguyen Pham
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000, Caen, France; Laboratoire de Physique Corpusculaire UMR6534 IN2P3/ENSICAEN, France - Normandie Université, France; Department of Radiation Oncology, Centre François Baclesse, Caen, Normandy, France
| | - Julie Coupey
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000, Caen, France
| | - Juliette Thariat
- Laboratoire de Physique Corpusculaire UMR6534 IN2P3/ENSICAEN, France - Normandie Université, France; Department of Radiation Oncology, Centre François Baclesse, Caen, Normandy, France
| | - Samuel Valable
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000, Caen, France.
| |
Collapse
|
16
|
Feng X, Liu X, Guan H, Chen C, Gao F, Gao X, Chen M, Zhao J, Xu Y, Wang M. Safety and Efficacy of Radiotherapy Combined With Sintilimab in Advanced NSCLC Patients Who Progressed on First or Second Line Therapy: A Prospective, Multiple Center, and Single-Arm Study. Thorac Cancer 2025; 16:e70043. [PMID: 40116232 PMCID: PMC11926647 DOI: 10.1111/1759-7714.70043] [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: 12/11/2024] [Revised: 03/03/2025] [Accepted: 03/09/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND This study explored the safety and efficacy of combining radiotherapy with sintilimab in non-small cell lung cancer (NSCLC) patients who have progressed after first or second-line therapy. METHODS In this multicenter, single-arm trial, patients with NSCLC who had progressed after first or second-line therapy were enrolled. Participants received hypofractionated stereotactic body radiotherapy (SBRT) (requiring a single-site biological dose of more than 30 Gy or planned to reach 30 Gy) followed by sintilimab every 3 weeks until disease progression or unacceptable toxicity occurred. RESULTS From March 1, 2019, to July 27, 2023, 14 patients were enrolled across two centers. The cohort included 64.3% males and 35.7% females, with a median age of 67 years (range 57-73 years). All participants completed radiation therapy and received at least one cycle of sintilimab. The overall response rate (ORR) was 21.4% (3/14) and the disease control rate (DCR) was 71.4% (10/14). The absent radiation response (ARR) was 14.3% (2/14). The median PFS was 4.17 months (95% CI: 1.15-8.69 months), with a 6-month PFS rate of 42.9%. The median OS was 16.17 months (95% CI: 11.69-20.64 months). Overall, 10 patients (71.4%) experienced at least one treatment-emergent adverse event (TEAE). Grade 3 adverse events included one case each of immune-related myocarditis, thrombocytopenia, and checkpoint inhibitor pneumonitis (CIP). Four patients (28.6%) had immune-related adverse events (irAEs) including skin rash and pruritus (2/14, grade 1), immune-related myocarditis (1/14, grade 3), and CIP (1/14, grade 3). CONCLUSIONS Radiotherapy combined with sintilimab for NSCLC patients who progressed after first-or second-line therapy showed promising efficacy outcomes.
Collapse
Affiliation(s)
- Xiaoyi Feng
- Department of Pulmonary and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Xiaoyan Liu
- Department of Pulmonary and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Hui Guan
- Department of RadiotherapyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Chunhong Chen
- Department of OncologyBeidahuang Group General HospitalHeilongjiangPeople's Republic of China
| | - Feng Gao
- Department of OncologyBeidahuang Group General HospitalHeilongjiangPeople's Republic of China
| | - Xiaoxing Gao
- Department of Pulmonary and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Minjiang Chen
- Department of Pulmonary and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Jing Zhao
- Department of Pulmonary and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Yan Xu
- Department of Pulmonary and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Mengzhao Wang
- Department of Pulmonary and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingPeople's Republic of China
| |
Collapse
|
17
|
Pan Y, Zhao H, Huang W, Liu S, Qi Y, Huang Y. Metal-Protein Hybrid Materials: Unlocking New Frontiers in Biomedical Applications. Adv Healthc Mater 2025; 14:e2404405. [PMID: 39778029 DOI: 10.1002/adhm.202404405] [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: 11/07/2024] [Revised: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Metal-protein hybrid materials represent a novel class of functional materials that exhibit exceptional physicochemical properties and tunable structures, rendering them remarkable applications in diverse fields, including materials engineering, biocatalysis, biosensing, and biomedicine. The design and development of multifunctional and biocompatible metal-protein hybrid materials have been the subject of extensive research and a key aspiration for practical applications in clinical settings. This review provides a comprehensive analysis of the design strategies, intrinsic properties, and biomedical applications of these hybrid materials, with a specific emphasis on their potential in cancer therapy, drug and vaccine delivery, antibacterial treatments, and tissue regeneration. Through rational design, stable metal-protein hybrid materials can be synthesized using straightforward methods, enabling them with therapeutic, delivery, immunomodulatory, and other desired functionalities. Finally, the review outlines the existing limitations and challenges associated with metal-protein hybrid materials and evaluates their potential for clinical translation, providing insights into their practical implementation within biomedical applications.
Collapse
Affiliation(s)
- Yong Pan
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Han Zhao
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Wenyong Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Siyang Liu
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yanxin Qi
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| |
Collapse
|
18
|
Peng P, Cao J, Cheng W, Ming H, He B, Duan X, Li L, Tian Y, Nice EC, Zhang Z, Huang C, Zheng S. Manganese dioxide-based in situ vaccine boosts antitumor immunity via simultaneous activation of immunogenic cell death and the STING pathway. Acta Biomater 2025; 194:467-482. [PMID: 39832699 DOI: 10.1016/j.actbio.2025.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
In situ vaccine (ISV) can activate the anti-tumor immune system by inducing immunogenic cell death (ICD) at the tumor site. However, the development of tumor ISV still faces challenges due to insufficient tumor antigens released by tumor cells and the existence of tumor immunosuppressive microenvironment (TIME). Targeting the STING pathway has been reported to enhance the adjuvant effects of in situ tumor vaccines by initiating innate immunity. Based on this, we developed a potent in situ cancer vaccine, MBMA-RGD ISV, which simultaneously induces ICD and activates the STING pathway to achieve sustained anti-tumor immunity. Specifically, a water-soluble prodrug Mit-ALA was synthesized from the chemotherapeutic agent mitoxantrone (Mit) and the photosensitizer precursor 5-aminolevulinic acid (5-ALA) by pH-responsive ester bonds, which was then loaded into pre-synthesized BSA-MnO2 nanoparticles and functionalized with the targeting Arg-Gly-Asp (RGD) peptide to obtain MBMA-RGD ISV. This ISV actively targets tumor cells by binding integrin receptors and then gradually releases antitumor components in response to tumor microenvironment (TME). The released 5-ALA is metabolized in mitochondria to produce photosensitizer PpIX. Under laser irradiation, the photodynamic property of PpIX coupled with the photothermal effect of Mit synergistically induced ICD, resulting in the release of tumor antigens and evoking adaptive immunity. Meanwhile, released Mn2+ and Mit synergistically activate the STING pathway by inducing DNA damage, further enhancing antitumor immunity. Moreover, large amounts of oxygen released by MnO2 relieved the hypoxia microenvironment, thus sensitizing photodynamic therapy and improving the immunosuppressive state of TME. Therefore, MBMA-RGD ISV efficiently activates systemic antitumor immunity in vitro and in vivo, providing new strategies and ideas for the development of tumor ISV. STATEMENT OF SIGNIFICANCE: Using a biocompatible BSA-MnO2 nanoplatform, we developed a dual-prodrug tumor in situ vaccine (ISV) with tumor microenvironment-responsive action for synergistic cancer immunotherapy. Once internalized by tumor cells, the MBMA-RGD ISV responded to intracellular H+, H2O2, and GSH, releasing its therapeutic "cargo." Under laser irradiation, the combined effects of photodynamic therapy (PDT) and photothermal therapy (PTT) induced immunogenic cell death (ICD), effectively recruiting and stimulating dendritic cells (DCs). Concurrently, STING pathway activation, triggered by DNA damage, enhanced DC maturation. Moreover, the MnO2 component alleviated hypoxia within the tumor microenvironment by releasing significant amounts of oxygen, which facilitated the repolarization of macrophages from the M2 phenotype to the M1 phenotype. Therefore, MBMA-RGD ISV demonstrated potent suppression of tumor metastasis and recurrence without notable side effects in mouse tumor models.
Collapse
Affiliation(s)
- Peilan Peng
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Jiangjun Cao
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Wenting Cheng
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Hui Ming
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Bo He
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Xirui Duan
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Lei Li
- Department of anorectal surgery, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yuan Tian
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Zhiqi Zhang
- Department of General Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
| | - Canhua Huang
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, China.
| | - Shaojiang Zheng
- Hainan Cancer Center and Tumor Institute, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China.
| |
Collapse
|
19
|
Baharom F, Hermans D, Delamarre L, Seder RA. Vax-Innate: improving therapeutic cancer vaccines by modulating T cells and the tumour microenvironment. Nat Rev Immunol 2025; 25:195-211. [PMID: 39433884 DOI: 10.1038/s41577-024-01091-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/23/2024]
Abstract
T cells have a critical role in mediating antitumour immunity. The success of immune checkpoint inhibitors (ICIs) for cancer treatment highlights how enhancing endogenous T cell responses can mediate tumour regression. However, mortality remains high for many cancers, especially in the metastatic setting. Based on advances in the genetic characterization of tumours and identification of tumour-specific antigens, individualized therapeutic cancer vaccines targeting mutated tumour antigens (neoantigens) are being developed to generate tumour-specific T cells for improved therapeutic responses. Early clinical trials using individualized neoantigen vaccines for patients with advanced disease had limited clinical efficacy despite demonstrated induction of T cell responses. Therefore, enhancing T cell activity by improving the magnitude, quality and breadth of T cell responses following vaccination is one current goal for improving outcome against metastatic tumours. Another major consideration is how T cells can be further optimized to function within the tumour microenvironment (TME). In this Perspective, we focus on neoantigen vaccines and propose a new approach, termed Vax-Innate, in which vaccination through intravenous delivery or in combination with tumour-targeting immune modulators may improve antitumour efficacy by simultaneously increasing the magnitude, quality and breadth of T cells while transforming the TME into a largely immunostimulatory environment for T cells.
Collapse
Affiliation(s)
| | - Dalton Hermans
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
| | | | - Robert A Seder
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
20
|
Zhang Q, Wang Q, Wang M, Liu X, Han D, Sun H, Zhao C, Liu C. Efficacy and safety of integrating consolidative thoracic radiotherapy with immunochemotherapy in extensive-stage small cell lung cancer: a real-world retrospective analysis. J Thorac Dis 2025; 17:836-848. [PMID: 40083492 PMCID: PMC11898347 DOI: 10.21037/jtd-24-1592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/20/2024] [Indexed: 03/16/2025]
Abstract
Background Extensive-stage small cell lung cancer (ES-SCLC) remains a challenging malignancy with a poor prognosis. The integration of immunochemotherapy and combined consolidative thoracic radiotherapy (cTRT) presents a potential paradigm shift in treatment. This study aims to evaluate the real-world efficacy and safety of this approach. Methods In a single-center retrospective study conducted at Shandong Cancer Hospital, electronic medical records of 828 ES-SCLC patients treated between January 1, 2022, and December 31, 2023, were reviewed. Patients were divided into three cohorts based on treatment strategies: chemoradiotherapy (cohort A), immunochemotherapy without/with cTRT (cohort B/C). Propensity score matching was utilized to adjust for baseline differences. The primary outcomes were real-world progression-free survival (rwPFS) and overall survival (OS). Secondary outcomes included the incidence and severity of specific interested adverse events (AEs). Results Of the 374 patients analyzed, cohort C showed significant improvements in rwPFS and OS compared to cohort A. The median rwPFS in cohort C (10.9 months) was longer than that of cohorts A (7.6 months) and B (8.0 months). The 12-month rwPFS rate was highest in cohort C (41%), compared to cohorts A (19%) and B (34%). The incidence of grade 3 or higher AEs was comparable across cohorts, with myelosuppression being the most common. However, the incidence of grade 3 or higher pneumonitis was notably higher in cohorts B and C, aligning with previous reports. Conclusions The combination of cTRT with immunochemotherapy for ES-SCLC showed improved rwPFS and OS, indicating potential benefit in this population. The overall safety profile remained manageable. These findings highlight the need for further prospective studies to confirm the optimal integration of cTRT in ES-SCLC treatment strategies.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Oncology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Qian Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Mengsen Wang
- Department of Oncology, Jining No. 1 People’s Hospital, Jining, China
| | - Xiaomeng Liu
- Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Dan Han
- Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Hongfu Sun
- Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Chengwei Zhao
- Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Chengxin Liu
- Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
21
|
Wei R, Xie K, Li T, Lin W, Zhao Y, Li J, Lai S, Wei X, Jiang X, Yuan Y, Yang R. Immunity/metabolism dual-regulation via an acidity-triggered bioorthogonal assembly nanoplatform enhances glioblastoma immunotherapy by targeting CXCL12/CXCR4 and adenosine-A2AR pathways. Biomaterials 2025; 319:123216. [PMID: 40037210 DOI: 10.1016/j.biomaterials.2025.123216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/16/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
Blocking the C-X-C motif chemokine ligand-12/C-X-C motif chemokine receptor-4 (CXCL12/CXCR4) signal offers the potential to induce immunogenic cell death (ICD) and enhance immunotherapy of glioblastoma (GBM). However, traditional intracellular targeted delivery strategies and adenosine-mediated tumor immunosuppression limit its therapeutic efficacy. Herein, we present an acidity-triggered self-assembly nanoplatform based on bioorthogonal reaction to potentiate GBM immunotherapy through dual regulation of metabolism and immune pathways. AMD3100 (CXCR4 antagonist) and CPI-444 (adenosine 2A receptor inhibitor) were formulated into micelles, denoted as AMD@iNPDBCO and CPI@iNPN3, respectively. Upon administration, the pH-sensitive poly(2-azepane ethyl methacrylate) group of AMD@iNPDBCO responds to the acidic tumor microenvironment, exposing the DBCO moiety, resulting in highly efficient bioorthogonal reaction with azide group on CPI@iNPN3 to form large-sized aggregates, ensuring extracellular drug release. The combination of AMD3100 and CPI-444 contributes to ICD induction, dendritic cell maturation, and immunosuppressive milieu alleviation by reducing tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells, leading to a robust antitumor response, thereby significantly prolonging survival in orthotopic GBM-bearing mice. Furthermore, the nanoplatform remarkably amplifies immuno-radiotherapy by potently evoking cytotoxic CD8+ T cell priming, and synergized with immune checkpoint blockade by delaying CD8+ T cell exhaustion. Our work highlights the potential of the in situ assembly nanoplatform tailored for delivery of extracellular-targeted therapeutic agents for boosting GBM immunotherapy.
Collapse
Affiliation(s)
- Ruili Wei
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Kunfeng Xie
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China
| | - Tao Li
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China
| | - Wanxian Lin
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Yandong Zhao
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Jiamin Li
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Shengsheng Lai
- School of Medical Equipment, Guangdong Food and Drug Vocational College, Guangzhou 510520, PR China
| | - Xinhua Wei
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Xinqing Jiang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Youyong Yuan
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China.
| | - Ruimeng Yang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China.
| |
Collapse
|
22
|
Liu W, Niu J, Huo Y, Zhang L, Han L, Zhang N, Yang M. Role of circular RNAs in cancer therapy resistance. Mol Cancer 2025; 24:55. [PMID: 39994791 PMCID: PMC11854110 DOI: 10.1186/s12943-025-02254-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Over the past decade, circular RNAs (circRNAs) have gained recognition as a novel class of genetic molecules, many of which are implicated in cancer pathogenesis via different mechanisms, including drug resistance, immune escape, and radio-resistance. ExosomalcircRNAs, in particular, facilitatecommunication between tumour cells and micro-environmental cells, including immune cells, fibroblasts, and other components. Notably, micro-environmental cells can reportedly influence tumour progression and treatment resistance by releasing exosomalcircRNAs. circRNAs often exhibit tissue- and cancer-specific expression patterns, and growing evidence highlights their potential clinical relevance and utility. These molecules show strong promise as potential biomarkers and therapeutic targets for cancer diagnosis and treatment. Therefore, this review aimed to briefly discuss the latest findings on the roles and resistance mechanisms of key circRNAs in the treatment of various malignancies, including lung, breast, liver, colorectal, and gastric cancers, as well as haematological malignancies and neuroblastoma.This review will contribute to the identification of new circRNA biomarkers for the early diagnosis as well as therapeutic targets for the treatment of cancer.
Collapse
Affiliation(s)
- Wenjuan Liu
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Jiling Niu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Yanfei Huo
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Long Zhang
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Linyu Han
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Nasha Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Ming Yang
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong Province, China.
| |
Collapse
|
23
|
Huang CJ, Liu GT, Yeh YC, Chung SY, Chang YC, Chiang NJ, Lu ML, Huang WN, Chen MH, Wang YC. Construction of hot tumor classification models in gastrointestinal cancers. J Transl Med 2025; 23:218. [PMID: 39984938 PMCID: PMC11846462 DOI: 10.1186/s12967-025-06230-x] [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: 11/18/2024] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Gastrointestinal (GI) cancers account for more than one-third of cancer-related mortality, and the prognosis for late-stage patients remains poor. Immunotherapy has been proven to extend the survival of patients at advanced stages; however, challenges persist in patient selection and overcoming drug resistance. Tumor-infiltrating lymphocytes (TILs) and tertiary lymphoid structures (TLS) in the tumor microenvironment (TME) have been found to be associated with anti-tumor immune responses. 'Hot tumors' with high levels of infiltration tend to respond better to immune checkpoint inhibitor (ICI) therapy, making them potential biomarkers for ICI treatment. METHODS To explore potential biomarkers for predicting immunotherapy response and prognosis in GI cancers, we downloaded the gene expression profiles of seven GI cancers from The Cancer Genome Atlas (TCGA) database and characterized their TME, classifying the samples into hot/cold tumor subgroups. Furthermore, we developed a computational framework to construct cancer-specific hot tumor classification models with only a few genes. External independent datasets and qPCR experiments were used to verify the performance of our few-gene models. RESULTS We constructed cancer-specific few-gene models to identify hot tumors for GI cancers with only two to nine genes. The results showed that B cells are important for hot tumor determination, and the identified hot tumors are significantly associated with TLS. They not only overexpress TLS marker genes but are also associated with the presence of TLS in whole-slide images. Further, a two-gene qPCR model was developed to effectively distinguish between hot and cold tumor subgroups in cholangiocarcinoma, providing an opportunity for stratifying patients with hot tumors in clinical settings. CONCLUSIONS In conclusion, our established few-gene models, which can be easily integrated into clinical practice, can distinguish hot and cold tumor subgroups, and may serve as potential biomarkers for predicting ICI response.
Collapse
Affiliation(s)
- Chien-Jung Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Guan-Ting Liu
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Chen Yeh
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shin-Yi Chung
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Nai-Jung Chiang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Meng-Lun Lu
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wei-Ning Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Huang Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Yu-Chao Wang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Digital Medicine and Smart Healthcare Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
24
|
Zhou W, Zhao Y, Qin W, Wu W, Liao C, Zhang Y, Yang X, Chen X, Wang Y, Kang Y, Wu J, Zhao J, Quan J, Wang X, Bu X, Yue X. Targeting USP1 Potentiates Radiation-Induced Type I IFN-Dependent Antitumor Immunity by Enhancing Oligo-Ubiquitinated SAR1A-Mediated STING Trafficking and Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412687. [PMID: 39976106 DOI: 10.1002/advs.202412687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/13/2025] [Indexed: 02/21/2025]
Abstract
The magnitude of Type I interferon (IFN) mediated innate immune response within the tumor microenvironment (TME) critically influences the effectiveness of radiotherapy. Unfortunately, due to a myriad of resistance mechanisms, the double-stranded DNA (dsDNA) signals produced by tumor cells postradiotherapy often induce a diminished response from immune cells. Through chemical screening targeting deubiquitinating enzymes, we identified USP1 (Ubiquitin Specific Peptidase 1) inhibitor as an enhancer of post-radiotherapy dsDNA responses. Mechanistically, within the context of immune-stimulatory cells in TME, USP1 serves as a suppressor in the stress-mediated stages of the cGAS (Cyclic GMP-AMP synthase) -STING (Stimulator of interferon genes protein) signaling pathway, specifically affecting the trafficking of STING from endoplasmic reticulum to Golgi apparatus. It is elucidated that SAR1A (Secretion associated Ras related GTPase 1A) requires K27-linked oligo-ubiquitination to assemble the STING-COP-II (Coat protein II) transport complex for STING trafficking. USP1 counteracts this activation by removing SAR1A ubiquitination, thereby blocking STING trafficking and activation. Consequently, pharmacological USP1 inhibition using ML323 sustains SAR1A ubiquitination and COP-II complex formation, significantly enhancing STING trafficking and subsequent Type I IFN production. This intervention substantially amplifies radiotherapy-induced immune activation in the TME, providing a strategic approach to overcome therapeutic resistance and synergize radiotherapy with immunotherapies.
Collapse
Affiliation(s)
- Weilin Zhou
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510006, China
| | - Yuxuan Zhao
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510006, China
| | - Wenjing Qin
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510630, China
| | - Weijian Wu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510006, China
| | - Chenyang Liao
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510006, China
| | - Yiqiu Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510006, China
| | - Xingli Yang
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xue Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510257, China
| | - Youqiao Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510006, China
| | - Yushan Kang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510006, China
| | - Jiaxin Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510257, China
| | - Jiaojiao Zhao
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510006, China
| | - Junmin Quan
- Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518072, China
| | - Xuecen Wang
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xianzhang Bu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510006, China
| | - Xin Yue
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510630, China
| |
Collapse
|
25
|
Rauf S, Smirnova A, Chang A, Liu Y, Jiang Y. Immunogenic Cell Death: the Key to Unlocking the Potential for Combined Radiation and Immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638342. [PMID: 40027799 PMCID: PMC11870562 DOI: 10.1101/2025.02.14.638342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Immunogenic cell death (ICD) enhances anti-tumor immunity by releasing tumor-associated antigens and activating the anti-tumor immune system response. However, its potential remains understudied in combination therapies. Here, we develop a mathematical model to quantify the role of ICD in optimizing the efficacy of combined radiotherapy (RT) and macrophage-based immunotherapy. Using preclinical murine data targeting the SIRP α -CD47 checkpoint, we show that RT alone induces minimal ICD, whereas disrupting the SIRP α -CD47 axis significantly enhances both phagocytosis and systemic immune activation. Our model predicts an optimal RT dose (6-8 Gy) for maximizing ICD, a dose-dependent abscopal effect, and a hierarchy of treatment efficacy, with SIRP α -knockout macrophages exhibiting the strongest tumoricidal activity. These findings provide a quantitative framework for designing more effective combination therapies, leveraging ICD to enhance immune checkpoint inhibition and radiotherapy synergy.
Collapse
|
26
|
Jiang Q, Chen Z, Jiang J, Chen Q, Lan H, Zhu J, Mao W. The role of cGAS-STING in remodeling the tumor immune microenvironment induced by radiotherapy. Crit Rev Oncol Hematol 2025; 209:104658. [PMID: 39956501 DOI: 10.1016/j.critrevonc.2025.104658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025] Open
Abstract
The activation of the cGAS-STING pathway occurs when tumor cell DNA is damaged by ionizing radiation. Once triggered, this pathway reshapes the tumor immune microenvironment by promoting the maturation, activation, polarization, and immune-killing capacity of immune cells, as well as by inducing the release of interferons and the expression of immune-related genes. In addition, the gut microbiota and various mechanisms of programmed cell death interact with the cGAS-STING pathway, further influencing its function in remodeling the immune microenvironment after radiotherapy. Therefore, investigating the mechanisms of the cGAS-STING pathway in reshaping the tumor immune microenvironment post-radiotherapy can not only optimize the efficacy of combined radiotherapy and immunotherapy but also provide new research directions and potential targets for cancer treatment.
Collapse
Affiliation(s)
- Qingyu Jiang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Zhejiang Chinese Medical University, Hangzhou 310053, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Zhiheng Chen
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing 31400, China
| | - Jin Jiang
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing 31400, China
| | - Qianping Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Huiyin Lan
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Ji Zhu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China.
| | - Wei Mao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China.
| |
Collapse
|
27
|
Zhang X, Shi C, Liu Q, Zhong Y, Zhu L, Zhao Y. Combination of adenosine blockade and ferroptosis for photo-immunotherapy of triple negative breast cancer with aptamer-modified copper sulfide. J Mater Chem B 2025; 13:2504-2519. [PMID: 39834279 DOI: 10.1039/d4tb02125h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Combination of immunotherapy and photothermal therapy (PTT) provides a promising therapeutic performance for tumors. However, it still faces negative feedback from suppressive factors such as adenosine. Herein, we developed a new nanodrug that can combine adenosine blockade and ferroptosis to promote the photoimmunotherapy of triple negative breast cancer (TNBC). The nanodrug, named CuS-PEG@Apt, was constructed via the modification of copper sulfide (CuS) nanoparticles with adenosine aptamer and PEG. CuS-PEG@Apt could be effectively enriched in the tumor site and locally generate a strong photothermal effect, directly ablating tumors and inducing immunogenic death (ICD). On the other hand, the aptamers could block the adenosine pathway to inhibit the immune suppression by adenosine, which further promoted the anti-tumor immunity. Moreover, the CuS nanoparticles could consume GSH and inhibit GPX4 to cause the ferroptosis of tumor cells. Collectively, CuS-PEG@Apt achieved potent efficacy of tumor suppression via the combination of PTT, immune activation and ferroptosis, representing an appealing platform for TNBC treatment.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Chengyu Shi
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Qiao Liu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Yuting Zhong
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Lipeng Zhu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Yuetao Zhao
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| |
Collapse
|
28
|
Ding Y, Feng Y, Ye Y, Shen J, Guo C, He X, Zhu L, Wang L. High and low dose radiotherapy combined with ICIs for MSS colorectal cancer patients with liver metastases: a phase I study (HaRyPOT). Front Oncol 2025; 15:1503517. [PMID: 39980556 PMCID: PMC11839429 DOI: 10.3389/fonc.2025.1503517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Microsatellite stable (MSS) colorectal cancer with liver metastases (CLRM) responds poorly to immunotherapy, and various approaches to break immune tolerance have been tried. Radiotherapy in combination with immune checkpoint inhibitors is one of promising therapies, and the choice of radiotherapy and immunotherapy modalities is also a hot issue. Methods Here, we report on a Phase I trial treating nine patients with MSS CLRM using a combination of high and low dose radiotherapy and immune checkpoint inhibitors (ICIs). Results The primary endpoint of the trial was the safety and tolerability of this combination treatment modality. Secondary endpoints included the objective response rate (ORR), progression-free survival (PFS) and overall survival (OS). The study results showed that at three dose levels-single doses of 6Gy (n=3), 8Gy (n=3), and 10Gy (n=3)-the most common treatment-related adverse events (TRAEs) were nausea, vomiting, fatigue, and abnormal liver function. At the first condition assessment, four patients were observed to have stable disease (SD) and one patient achieved partial response (PR). In exploratory endpoint analyses, tissue biopsies and paired hematologic samples from patients showed M2 macrophage reduction. Plasma cytokines IL-10, IL-17, and INF-α increased after treatment with both drugs. Discussion In summary, this is the first clinical trial demonstrating the safety and immunogenic activity of combined high and low dose radiotherapy with ICIs in MSS colorectal cancer liver metastases (CRLMs). The combination therapy stimulated the immune response and altered the tumour microenvironment, warranting further exploration in the future.
Collapse
Affiliation(s)
- Yuxuan Ding
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yong Feng
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yangfan Ye
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiayi Shen
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Chang Guo
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xia He
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Liangjun Zhu
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Lijun Wang
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| |
Collapse
|
29
|
Yu J, Yin L, Guo W, Wang Q, Liu J, Zhang L, Ye H, Xia J, Xia Y, Wu J, Wang W, Yang Y, Zong D, He X, Wang L, Jiang H. Hypofractionated radiotherapy combined with a PD-1 inhibitor, granulocyte macrophage-colony stimulating factor, and thymosin-α1 in advanced metastatic solid tumors: a multicenter Phase II clinical trial. Cancer Immunol Immunother 2025; 74:98. [PMID: 39904914 PMCID: PMC11794727 DOI: 10.1007/s00262-024-03934-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/27/2024] [Indexed: 02/06/2025]
Abstract
PURPOSE This multicenter Phase II clinical study assessed the efficacy and safety of hypofractionated radiotherapy (HFRT) in combination with a PD-1 inhibitor, granulocyte macrophage-colony stimulating factor (GM-CSF), and thymosin-α1 in patients with heavily treated metastatic solid tumors. METHODS Patients were enrolled between September 2022 and May 2024. HFRT was administered to targeted tumors, and GM-CSF was administered for 14 days from day 1 of radiotherapy. Thymosin-α1 was injected concurrently twice weekly until disease progression. Immunotherapy with camrelizumab was started following HFRT and repeated every 3 weeks. GM-CSF was administered daily for 7 days before each cycle of immunotherapy. RESULTS By June 15, 2024, there were 37 study participants. The median follow-up duration was 5.97 months (range 0.40-20.9). Median progression-free survival was 3.5 months (95% confidence interval 2.73-4.23) in the intention-to-treat population. The objective response rate was 23.08%, and the disease control rate was 65.38%. Overall survival data are not yet mature. Abscopal effects were observed in 6 patients (23.08%); four of whom achieved a partial response. Patients who achieved a partial response were significantly more likely to have an abscopal effect( P = 0.025). The group with a lower baseline neutrophil-lymphocyte ratio had a significantly lower risks of distant metastasis and death( P = 0.024). Seventeen adverse reactions were reported, including six grade 3 or 4 adverse events. There were no grade 5 adverse events. CONCLUSION In conclusion, the trends in efficacy observed in our study are promising; however, well-designed protocols are essential to validate these findings.
Collapse
Affiliation(s)
- Jiamin Yu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Nanjing Medical University, &Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, China
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Li Yin
- Department of Radiation Oncology, Affiliated Cancer Hospital of Nanjing Medical University, &Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - Wenjie Guo
- Department of Radiation Oncology, Affiliated Cancer Hospital of Nanjing Medical University, &Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - Qiang Wang
- Department of Radiation Oncology, Xuzhou Cancer Hospital, Xuzhou, 221005, China
| | - Juying Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Nanjing Medical University, &Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - Lansheng Zhang
- Department of Radiation Oncology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| | - Hongxun Ye
- Department of Radiation Oncology, Taixing People's Hospital Affiliated With Medical College of Yangzhou University, Taizhou, 225400, China
| | - Jianhong Xia
- Department of Radiation Oncology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, China
| | - Youyou Xia
- Department of Radiation Oncology, The First People's Hospital of Lianyungang, Lianyungang, 222061, China
| | - Jianfeng Wu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Nanjing Medical University, &Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - Wanwei Wang
- Department of Radiation Oncology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Yanguang Yang
- Department of Radiation Oncology, Nantong Cancer Hospital, Nantong, 226361, China
| | - Dan Zong
- Department of Radiation Oncology, Affiliated Cancer Hospital of Nanjing Medical University, &Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - Xia He
- Department of Radiation Oncology, Affiliated Cancer Hospital of Nanjing Medical University, &Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, China.
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Lijun Wang
- Department of Radiation Oncology, Affiliated Cancer Hospital of Nanjing Medical University, &Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, China.
| | - Hong Jiang
- Department of Radiation Oncology, The People's Hospital of Jiawang District of Xuzhou City, Xuzhou, 221011, China.
| |
Collapse
|
30
|
Yesmembetov K, Sahin C, Murad M, Berres ML, Koch A, von Websky M, Vondran F, Bruners P, Eble M, Mohamed AA. Potential radiotherapy-related reactivation of immune checkpoint inhibitor hepatitis. Strahlenther Onkol 2025:10.1007/s00066-024-02361-0. [PMID: 39904779 DOI: 10.1007/s00066-024-02361-0] [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: 07/10/2024] [Accepted: 12/23/2024] [Indexed: 02/06/2025]
Abstract
This report details the reactivation of immune checkpoint inhibitor (ICI)-related autoimmune hepatitis triggered by stereotactic body radiation therapy (SBRT) in a 55-year-old male with hilar cholangiocellular carcinoma. Initially diagnosed in December 2021, the patient underwent successful resection and subsequent adjuvant therapy. Despite stable disease following chemotherapy augmented with durvalumab, he developed grade 3 acute hepatitis after seven cycles of durvalumab. Following a brief prednisolone regimen and normalization of liver tests, SBRT targeting para-aortic lymph nodes was initiated. Remarkably, severe hepatitis reoccurred 7 days after starting SBRT, 88 days following the last durvalumab infusion, necessitating resumed and escalated prednisolone treatment. Another course of SBRT for a newly diagnosed metastatic liver lesion was administered in September 2023, with ongoing prednisolone adjustment. By February 2024, liver tests normalized, but subsequent radiological assessments indicated tumor progression, leading to the reintroduction of chemotherapy. This case underscores the potential of SBRT for activating severe immune-mediated hepatotoxicity in patients treated with ICIs, highlighting the need for careful monitoring and management of such patients. Further, this report highlights the possible survival benefit of the strategic application of SBRT in addition to systematic treatment in recurrent and metastatic cholangiocellular carcinoma.
Collapse
Affiliation(s)
- Kakharman Yesmembetov
- Gastroenterology, Hepatology and infectious Diseases Department, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen, Bonn, Cologne and Duesseldorf (CIO ABCD), Aachen, Germany
- National research oncology center, Center for Hepatopancreatobiliary Surgery and Organ Transplantation, Kerey Zhanibek khandar street 3, Z05K4F3, Astana, Kazakhstan
| | - Cennet Sahin
- Gastroenterology, Hepatology and infectious Diseases Department, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen, Bonn, Cologne and Duesseldorf (CIO ABCD), Aachen, Germany
| | - Mohamad Murad
- Gastroenterology, Hepatology and infectious Diseases Department, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen, Bonn, Cologne and Duesseldorf (CIO ABCD), Aachen, Germany
| | - Marie-Luise Berres
- Gastroenterology, Hepatology and infectious Diseases Department, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen, Bonn, Cologne and Duesseldorf (CIO ABCD), Aachen, Germany
| | - Alexander Koch
- Gastroenterology, Hepatology and infectious Diseases Department, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen, Bonn, Cologne and Duesseldorf (CIO ABCD), Aachen, Germany
| | - Martin von Websky
- Visceral and Transplantation Surgery Department, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen, Bonn, Cologne and Duesseldorf (CIO ABCD), Aachen, Germany
| | - Florian Vondran
- Visceral and Transplantation Surgery Department, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen, Bonn, Cologne and Duesseldorf (CIO ABCD), Aachen, Germany
| | - Philipp Bruners
- Diagnostic and interventional Radiology Department, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen, Bonn, Cologne and Duesseldorf (CIO ABCD), Aachen, Germany
| | - Michael Eble
- Radiation Oncology Department, University Hospital RWTH Aachen, Pauwelstraße 30, 52074, Aachen, Germany
- Center for Integrated Oncology Aachen, Bonn, Cologne and Duesseldorf (CIO ABCD), Aachen, Germany
| | - Ahmed Allam Mohamed
- Radiation Oncology Department, University Hospital RWTH Aachen, Pauwelstraße 30, 52074, Aachen, Germany.
- Center for Integrated Oncology Aachen, Bonn, Cologne and Duesseldorf (CIO ABCD), Aachen, Germany.
| |
Collapse
|
31
|
Misawa K, Bhat H, Adusumilli PS, Hou Z. Combinational CAR T-cell therapy for solid tumors: Requisites, rationales, and trials. Pharmacol Ther 2025; 266:108763. [PMID: 39617146 PMCID: PMC11848936 DOI: 10.1016/j.pharmthera.2024.108763] [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: 09/06/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved potent antitumor efficacy in hematological malignancies; however, because of limitations in CAR T-cell recruitment, infiltration, activation, and functional persistence in the tumor, its efficacy in solid tumors has been suboptimal. To overcome these challenges, combinational strategies that include chemotherapy, radiation therapy, or immune checkpoint inhibitor agent therapy with CAR T-cell therapy are being investigated. The established functional characteristics of the abovementioned therapies provide a rationale for the use of a combinational approach with CAR T cells. Chemotherapy reshapes the peritumoral stroma, decreases the immunosuppressive cell population, and promotes a proinflammatory milieu, all of which allow for increased recruitment, infiltration, and accumulation of CAR T cells. Radiation therapy promotes a chemokine gradient, which augments tumor infiltration by CAR T cells and further increases expression of tumor-associated antigens, allowing for increased activation of CAR T cells. Immune checkpoint inhibitor agent therapy inactivates T-cell exhaustion pathways-most notably, the PD1/PDL1 pathway-thereby improving the functional persistence of CAR T cells and promoting endogenous immunity. In this review, we discuss the requisites and rationales for combinational therapy, and we review 25 ongoing phase I and II clinical trials, of which 4 use chemotherapy, 3 use radiation therapy, 11 use immunotherapy, and 7 use another agent. While safety, efficacy, and improved outcomes are the primary goals of these ongoing studies, the knowledge gained from them will help pave the way for subsequent studies focused on optimizing combinational regimens and identifying predictive biomarkers.
Collapse
Affiliation(s)
- Kyohei Misawa
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Hina Bhat
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Zhaohua Hou
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
32
|
Liang J, Ma M, Feng W, Xu Q, Chen D, Lai J, Chen J. Anti-PD-L1 blockade facilitates antitumor effects of radiofrequency ablation by improving tumor immune microenvironment in hepatocellular carcinoma. Apoptosis 2025; 30:55-68. [PMID: 39327353 PMCID: PMC11799020 DOI: 10.1007/s10495-024-02019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 09/28/2024]
Abstract
Hepatocellular carcinoma (HCC) is a complex disease with advanced presentation that significantly affects survival rates. Therefore, novel therapeutic strategies are needed. In this study, we investigate the tumor microenvironment (TME) in HCC by analyzing 13 HCC samples at single cell level. We identified key cell populations, including CD8 + T cells, Tregs, M1/M2 macrophages, and CD4 + memory T cells, and explored their roles and interactions. Our research revealed an early enrichment of CD8 + T cells, which could potentially lead to their exhaustion and facilitate tumor progression. We also investigated the impact of percutaneous radiofrequency ablation (RFA) on the immune microenvironment. Using a dual tumor mouse model, we demonstrated that RFA induces necrosis, enhancing antigen presentation and altering immune responses. Our results indicate that RFA increases PD-L1 expression in residual liver tissue, suggesting potential immune escape mechanisms. Furthermore, the combination of RFA and anti-PD-L1 therapy in the mouse model resulted in significant improvements in immune modulation. This included increased CD8 + T cell efficacy and decreased Treg infiltration. This combination shows promise as an approach to counteract HCC progression by altering the immune landscape. This study highlights the critical interaction within the TME of HCC and suggests the possibility of improving patient outcomes by targeting immune evasion mechanisms through combined therapeutic strategies.
Collapse
Affiliation(s)
- Jiahua Liang
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Medicine II, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, Mannheim, Germany
| | - Mingjian Ma
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wei Feng
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qiongcong Xu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Dong Chen
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiaming Lai
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Jiancong Chen
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
33
|
Xiong Y, Li J, Jiang X, Zhen W, Ma X, Lin W. Nitric Oxide-Releasing Nanoscale Metal-Organic Layer Overcomes Hypoxia and Reactive Oxygen Species Diffusion Barriers to Enhance Cancer Radiotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413518. [PMID: 39742392 PMCID: PMC11848595 DOI: 10.1002/advs.202413518] [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: 10/23/2024] [Revised: 12/17/2024] [Indexed: 01/03/2025]
Abstract
Hafnium (Hf)-based nanoscale metal-organic layers (MOLs) enhance radiotherapeutic effects of tissue-penetrating X-rays via a unique radiotherapy-radiodynamic therapy (RT-RDT) process through efficient generation of hydroxy radical (RT) and singlet oxygen (RDT). However, their radiotherapeutic efficacy is limited by hypoxia in deep-seated tumors and short half-lives of reactive oxygen species (ROS). Herein the conjugation of a nitric oxide (NO) donor, S-nitroso-N-acetyl-DL-penicillamine (SNAP), to the Hf12 secondary building units (SBUs) of Hf-5,5'-di-p-benzoatoporphyrin MOL is reported to afford SNAP/MOL for enhanced cancer radiotherapy. Under X-ray irradiation, SNAP/MOL efficiently generates superoxide anion (O2 -.) and releases nitric oxide (NO) in a spatio-temporally synchronized fashion. The released NO rapidly reacts with O2 -. to form long-lived and highly cytotoxic peroxynitrite which diffuses freely to the cell nucleus and efficiently causes DNA double-strand breaks. Meanwhile, the sustained release of NO from SNAP/MOL in the tumor microenvironment relieves tumor hypoxia to reduce radioresistance of tumor cells. Consequently, SNAP/MOL plus low-dose X-ray irradiation efficiently inhibits tumor growth and reduces metastasis in colorectal and triple-negative breast cancer models.
Collapse
Affiliation(s)
- Yuxuan Xiong
- Department of ChemistryThe University of ChicagoChicagoIL60637USA
| | - Jinhong Li
- Department of ChemistryThe University of ChicagoChicagoIL60637USA
| | - Xiaomin Jiang
- Department of ChemistryThe University of ChicagoChicagoIL60637USA
| | - Wenyao Zhen
- Department of ChemistryThe University of ChicagoChicagoIL60637USA
| | - Xin Ma
- Department of ChemistryThe University of ChicagoChicagoIL60637USA
| | - Wenbin Lin
- Department of ChemistryThe University of ChicagoChicagoIL60637USA
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis ResearchThe University of ChicagoChicagoIL60637USA
| |
Collapse
|
34
|
Yang W, Di S, Yang Z, Cao J, Fu Q, Ren H, Cheng H, Xie Y, Jia W, Dai X, Yu M, Chen Y, Cui X. One-dimensional nanosonosensitizer boosted multiple branches of immune responses against MHC-deficient immune-evasive urologic tumor. SCIENCE ADVANCES 2025; 11:eado7373. [PMID: 39879294 PMCID: PMC11777198 DOI: 10.1126/sciadv.ado7373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 08/26/2024] [Indexed: 01/31/2025]
Abstract
Cancer immunotherapies rely on CD8+ cytolytic T lymphocytes (CTLs) in recognition and eradication of tumor cells via antigens presented on major histocompatibility complex class I (MHC-I) molecules. However, we observe MHC-I deficiency in human and murine urologic tumors, posing daunting challenges for successful immunotherapy. We herein report an unprecedented nanosonosensitizer of one-dimensional bamboo-like multisegmented manganese dioxide@manganese-bismuth vanadate (BMMBV) to boost multiple branches of immune responses targeting MHC-I-deficient tumors. BMMBV markedly augments sonodynamic activity contributed by manganese heteroatoms in the lattice of bismuth vanadate with narrowing bandgaps. Under sonoirradiation, BMMBV enhances tumor antigen spreading and emission of adjuvant signals, which potentiate dendritic cell maturation, thereby eliciting high aptitude of CTLs. This therapy substantially up-regulates MHC expression on tumor cells, which are reversely sensitive to CTLs. Alongside, extensive innate immune cells complement the cytolytic activity of CTLs for eliminating mouse urologic tumors. This study offers a reinforced strategy against antigen-loss immune-evasive tumor.
Collapse
Affiliation(s)
- Wei Yang
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, P. R. China
| | - Sichen Di
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, P. R. China
| | - Zihuan Yang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Jianwei Cao
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, P. R. China
| | - Qingqiao Fu
- School of Materials Science and Engineering, Shanghai University, Shanghai 200444, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Hui Cheng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Xinyue Dai
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang 325088, P. R. China
- Shanghai Institute of Materdicine, Shanghai 200051, P. R. China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, P. R. China
| |
Collapse
|
35
|
Luna-Gutiérrez M, Azorín-Vega E, Oros-Pantoja R, Ocampo-García B, Cruz-Nova P, Jiménez-Mancilla N, Bravo-Villegas G, Santos-Cuevas C, Meléndez-Alafort L, Ferro-Flores G. Lutetium-177 labeled iPD-L1 as a novel immunomodulator for cancer-targeted radiotherapy. EJNMMI Radiopharm Chem 2025; 10:5. [PMID: 39843795 PMCID: PMC11754567 DOI: 10.1186/s41181-025-00328-9] [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: 12/02/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Cancer immunotherapy is a relatively new approach to cancer treatment. Peptides that target specific pathways and cells involved in immunomodulation can potentially improve the efficacy of cancer therapy. Recently, we reported iPD-L1 as a novel inhibitor peptide that specifically targets the cancer cell ligand PD-L1 (programmed death ligand 1). PD-L1 is responsible for inhibiting the immune checkpoint protein PD-1 expressed by regulatory T cells. On the other hand, anti-PD-L1 immunotherapy in combination with external beam radiotherapy has shown improved outcomes in the treatment of breast and lung cancer. The aim of this research was to prepare 177Lu-labeled iPD-L1 and to preclinically evaluate its radiotherapeutic potential and role as a tumor immunomodulator by measuring macrophage activation, IL-10, TGFβ, and PD-L1 expression in 4T1 triple-negative breast cancer cells and murine 4T1 tumors after treatment with 177Lu-iPD-L1. RESULTS The iPD-L1 ligand, characterized by UPLC mass, UV-Vis, and FT-IR spectroscopies, showed a chemical purity of 99%. The 177Lu-iPD-L1 radiochemical purity was 98.9 ± 1.1%. In vitro and in vivo studies demonstrated radiotracer stability in human serum (> 97% after 24 h evaluated by radio-HPLC), adequate affinity by the PDL1 protein (IC50 = 4.21 nM), and specific detection for PD-L1 assessed in 4T1, HCT116, and AR42J cancer cells, in which PD-L1 expression was verified by immunofluorescence and Western Blot assays. After treatment with 177Lu-iPD-L1 (0.4 Bq/cell), flow cytometry results showed a significant decrease in cell viability of 4T1 cells (dead 56.2%) compared to 177LuCl3 (dead 34.2%) and untreated cells (dead 9.4%). With high tumor uptake (6.97 ± 1.04%ID) and hepatobiliary and renal clearance, lutetium-177-labeled iPD-L1 delivered a tumor dose of 27 Gy/37 MBq and less than 0.36 Gy/37 MBq to non-source organs. PD-L1 positive tumors showed a significant increase in activated macrophages, PD-L1, IL-10, and TGFβ expression levels after 177Lu-iPD-L1 treatment as evaluated by ELISA assay and immunohistochemistry. CONCLUSIONS Therefore, this study warrants further dosimetric and clinical studies to determine the immunomodulatory effect and therapeutic efficacy of 177Lu-iPD-L1 in treating PD-L1-positive tumors in combination with anti-PD-1/PD-L1 immunotherapy protocols.
Collapse
Affiliation(s)
- Myrna Luna-Gutiérrez
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, 52750, Mexico
| | - Erika Azorín-Vega
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, 52750, Mexico
| | | | - Blanca Ocampo-García
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, 52750, Mexico
| | - Pedro Cruz-Nova
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, 52750, Mexico
| | | | | | - Clara Santos-Cuevas
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, 52750, Mexico
| | - Laura Meléndez-Alafort
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, 35128, Italy.
| | - Guillermina Ferro-Flores
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, 52750, Mexico.
| |
Collapse
|
36
|
Iwamoto FM, Tanguturi SK, Nayak L, Wang TJ, Desai A, Lustig RA, Bagley S, Wong ET, Hertan LM, McCluskey C, Hayden J, Muzikansky A, Nakhawa S, Japo J, Bossi CC, Meylan M, Tian Y, Barlow GL, Speliakos P, Ayoub G, Meredith DM, Ligon KL, Haas-Kogan D, Huang K, Wucherpfennig KW, Wen PY, Reardon DA. Re-Irradiation Plus Pembrolizumab: A Phase II Study for Patients with Recurrent Glioblastoma. Clin Cancer Res 2025; 31:316-327. [PMID: 39513953 DOI: 10.1158/1078-0432.ccr-24-1629] [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: 06/02/2024] [Revised: 07/15/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Radiotherapy may enhance antitumor immune responses by several mechanisms, including induction of immunogenic cell death. We performed a phase 2 study of pembrolizumab with re-irradiation in patients with recurrent glioblastoma. PATIENTS AND METHODS Sixty patients with recurrent glioblastoma received pembrolizumab with re-irradiation alone (cohort A, bevacizumab-naïve; n = 30) or with bevacizumab continuation (cohort B, n = 30). Dual primary endpoints, including the overall response rate and overall survival (OS) at either 12 (OS-12; cohort A) or 6 months (OS-6; cohort B), were assessed per cohort relative to historic benchmarks. Paired paraffin-embedded formalin-fixed tumor samples were assessed for immunologic biomarkers by IHC using digital quantification and co-detection-by-indexing (CODEX). RESULTS Study therapy was well tolerated, with most toxicities being grade ≤3. For cohort B, the primary endpoint of OS-6 was achieved (57%); however, survival was not improved for cohort A patients. The overall response rate was 3.3% and 6.7% for cohorts A and B, respectively. CODEX analysis of paired tumor samples from five patients revealed an increase of activated T cells in the tumor microenvironment after study therapy. CONCLUSIONS Compared with historic controls, re-irradiation plus pembrolizumab seemed to improve survival among bevacizumab-refractory patients but not among bevacizumab-naïve patients. CODEX revealed evidence of intratumoral infiltration of activated immune effector cells. A randomized, properly controlled trial of PD-1 blockade plus re-irradiation is warranted to further evaluate this regimen for bevacizumab-refractory patients, but alternative approaches are needed for bevacizumab-naïve patients.
Collapse
MESH Headings
- Humans
- Glioblastoma/therapy
- Glioblastoma/pathology
- Glioblastoma/drug therapy
- Glioblastoma/mortality
- Glioblastoma/immunology
- Male
- Female
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Middle Aged
- Aged
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/immunology
- Adult
- Re-Irradiation
- Brain Neoplasms/therapy
- Brain Neoplasms/immunology
- Brain Neoplasms/drug therapy
- Brain Neoplasms/pathology
- Brain Neoplasms/mortality
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Bevacizumab/administration & dosage
- Bevacizumab/therapeutic use
- Combined Modality Therapy
- Treatment Outcome
Collapse
Affiliation(s)
- Fabio M Iwamoto
- Department of Neurology, Columbia University Medical Center, New York, New York
| | - Shyam K Tanguturi
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lakshmi Nayak
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tony J Wang
- Department of Radiation Oncology, Columbia University Medical Center, New York, New York
| | - Arati Desai
- Department of Medical Oncology, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Robert A Lustig
- Department of Radiation Oncology, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Stephen Bagley
- Department of Medical Oncology, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Eric T Wong
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Lauren M Hertan
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Christine McCluskey
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Julia Hayden
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alona Muzikansky
- Department of Biostatistics, Massachusetts General Hospital, Boston, Massachusetts
| | - Shreya Nakhawa
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Julia Japo
- Department of Neuropathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Connor C Bossi
- Department of Neuropathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Maxime Meylan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ye Tian
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Graham L Barlow
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Paul Speliakos
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Georges Ayoub
- Department of Neuropathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David M Meredith
- Department of Neuropathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Keith L Ligon
- Department of Neuropathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kun Huang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
37
|
Zhan S, Cao Z, Li J, Chen F, Lai X, Yang W, Teng Y, Li Z, Zhang W, Xie J. Iron Oxide Nanoparticles Induce Macrophage Secretion of ATP and HMGB1 to Enhance Irradiation-Led Immunogenic Cell Death. Bioconjug Chem 2025; 36:80-91. [PMID: 39680043 PMCID: PMC11740999 DOI: 10.1021/acs.bioconjchem.4c00488] [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: 10/26/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
ATP (adenosine triphosphate) and HMGB1 (high mobility group box 1 protein) are key players in treatments that induce immunogenic cell death (ICD). However, conventional therapies, including radiotherapy, are often insufficient to induce ICD. In this study, we explore a strategy using nanoparticle-loaded macrophages as a source of ATP and HMGB1 to complement radiation-induced intrinsic and adaptive immune responses. To this end, we tested three inorganic particles, namely, iron oxide nanoparticles (ION), aluminum oxide nanoparticles (AON), and zinc oxide nanoparticles (ZON), in vitro with bone marrow-derived dendritic cells (BMDCs) and then in vivo in syngeneic tumor models. Our results showed that ION was the most effective of the three nanoparticles in promoting the secretion of ATP and HMGB1 from macrophages without negatively affecting macrophage survival. Secretions from ION-loaded macrophages can activate BMDCs. Intratumoral injection of ION-loaded macrophages significantly enhanced tumor infiltration and activation of dendritic cells and cytotoxic T cells. Moreover, exogenous ION macrophages can enhance the efficacy of radiotherapy. In addition, direct injection of ION can also enhance the efficacy of radiotherapy, which is attributed to ION uptake by and stimulation of endogenous macrophages. Instead of directly targeting cancer cells, our strategy targets macrophages and uses them as a secretory source of ATP and HMGB1 to enhance radiation-induced ICD. Our research introduces a new nanoparticle-based immunomodulatory approach that may have applications in radiotherapy and beyond.
Collapse
Affiliation(s)
- Shuyue Zhan
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Zhengwei Cao
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Jianwen Li
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Fanghui Chen
- Department
of Hematology and Medical Oncology & Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Xinning Lai
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Wei Yang
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Yong Teng
- Department
of Hematology and Medical Oncology & Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Zibo Li
- Department
of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive
Cancer Center, University of North Carolina
at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Weizhong Zhang
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Jin Xie
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
38
|
Russell SN, Demetriou C, Valenzano G, Evans A, Go S, Stanly T, Hazini A, Willenbrock F, Gordon-Weeks AN, Mukherjee S, Tesson M, Morton JP, O'Neill E, Jones KI. Induction of macrophage efferocytosis in pancreatic cancer via PI3Kγ inhibition and radiotherapy promotes tumour control. Gut 2025:gutjnl-2024-333492. [PMID: 39788719 DOI: 10.1136/gutjnl-2024-333492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND The immune suppression mechanisms in pancreatic ductal adenocarcinoma (PDAC) remain unknown, but preclinical studies have implicated macrophage-mediated immune tolerance. Hence, pathways that regulate macrophage phenotype are of strategic interest, with reprogramming strategies focusing on inhibitors of phosphoinositide 3-kinase-gamma (PI3Kγ) due to restricted immune cell expression. Inhibition of PI3Kγ alone is ineffective in PDAC, despite increased infiltration of CD8+ T cells. OBJECTIVE We hypothesised that the immune stimulatory effects of radiation, and its ability to boost tumour antigen availability could synergise with PI3Kγ inhibition to augment antitumour immunity. DESIGN We used orthoptic and genetically engineered mouse models of pancreatic cancer (LSL-KrasG12D/+;Trp53R172H/+;Pdx1-Cre). Stereotactic radiotherapy was delivered using contrast CT imaging, and PI3Kγ inhibitors by oral administration. Changes in the tumour microenvironment were quantified by flow cytometry, multiplex immunohistochemistry and RNA sequencing. Tumour-educated macrophages were used to investigate efferocytosis, antigen presentation and CD8+ T cell activation. Single-cell RNA sequencing data and fresh tumour samples with autologous macrophages to validate our findings. RESULTS Tumour-associated macrophages that employ efferocytosis to eradicate apoptotic cells can be redirected to present tumour antigens, stimulate CD8+ T cell responses and increase local tumour control. Specifically, we demonstrate how PI3Kγ signalling restricts inflammatory macrophages and that inhibition supports MERTK-dependent efferocytosis. We further find that the combination of PI3Kγ inhibition with targeted radiotherapy stimulates inflammatory macrophages to invoke a pathogen-induced like efferocytosis that switches from immune tolerant to antigen presenting. CONCLUSIONS Our data supports a new immunotherapeutic approach and a translational rationale to improve survival in PDAC.
Collapse
Affiliation(s)
| | | | | | - Alice Evans
- Department of Oncology, University of Oxford, Oxford, UK
| | - Simei Go
- Department of Oncology, University of Oxford, Oxford, UK
| | - Tess Stanly
- Department of Oncology, University of Oxford, Oxford, UK
| | - Ahmet Hazini
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | | | - Matthias Tesson
- Institute of Cancer Sciences, CRUK Scotland Institute, Glasgow, UK
| | | | - Eric O'Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | - Keaton Ian Jones
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
39
|
Harthorn A, Kuo TH, Torres SW, Lobb RR, Hackel BJ. Expression-Dependent Tumor Pretargeting via Engineered Avidity. Mol Pharm 2025; 22:558-572. [PMID: 39704255 DOI: 10.1021/acs.molpharmaceut.4c01177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Selective delivery of therapeutic modalities to tumor cells via binding of tumor-selective cell-surface biomarkers has empowered substantial advances in cancer treatment. Yet, tumor cells generally lack a truly specific biomarker that is present in high density on tumor tissue while being completely absent from healthy tissue. Rather, low but nonzero expression in healthy tissues results in on-target, off-tumor activity with detrimental side effects that constrain the therapeutic window or prevent use altogether. Advanced technologies to enhance the selectivity for tumor targeting are sorely needed. We have engineered a binding platform that is quantitatively dependent upon expression levels, via avidity-driven specificity, rather than binarily reliant on the presence or absence of a biomarker. We systematically varied monomeric binding affinity by engineering affibodies to target carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) and folate receptor 1 (FolR1). Two identical affibody ligands were tethered, with varying polypeptide linker lengths, to a nanobody that binds Alfa peptide to create a bispecific, trivalent protein for use in pretargeted radioligand therapy. Expression-dependent targeting was achieved in both systems: with 110 nM monomeric affinity to CEACAM5 with a two-amino-acid linker or with 250 nM monomeric affinity for FolR1 and a 10 amino acid linker. The latter bispecific, trivalent achieved over 25-fold differentiation between FolR1high and FolR1low cells in a mixed culture. Similar selectivity was achieved in a size-efficient bivalent molecule lacking a central nanobody. Moreover, the avid bivalent affibody molecule exhibited minimal inhibition by soluble antigen, whereas high-affinity bivalent antibody was inhibited by 97 ± 2%, which is indicative of serum inhibition of shed antigen. This work advances design principles for achieving expression-dependent tumor targeting via low-affinity, high-avidity ligands.
Collapse
Affiliation(s)
- Abbigael Harthorn
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Tse-Han Kuo
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Sarah W Torres
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Roy R Lobb
- MRB Biotherapeutics, Newton Center, Massachusetts 02459, United States
| | - Benjamin J Hackel
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
- MRB Biotherapeutics, Newton Center, Massachusetts 02459, United States
| |
Collapse
|
40
|
Uboha NV, Basree MM, Eickhoff JC, Deming DA, Matkowskyj K, Maloney J, McCarthy D, DeCamp M, LoConte N, Emmerich PB, Kraus S, Patel MA, Kratz JD, Lubner SJ, Hurst N, Bassetti MF. Phase I/II Trial of Perioperative Avelumab in Combination With Chemoradiation in the Treatment of Stage II/III Resectable Esophageal and Gastroesophageal Junction Cancer. J Surg Oncol 2025. [PMID: 39757733 DOI: 10.1002/jso.28070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/18/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND AND OBJECTIVES Standard treatment of patients with stage II/III esophageal or gastroesophageal junction (E/GEJ) cancer involves neoadjuvant chemoradiation (nCRT), resection, and immunotherapy. Our trial evaluated the addition of perioperative avelumab to standard treatments. METHODS Patients with resectable E/GEJ cancers received avelumab with nCRT and adjuvant avelumab after resection. Primary endpoints for phase I and II portions were safety and pathologic complete response (pCR) rate, respectively. Secondary endpoints included recurrence-free survival (RFS), surgical complication prevalence, and R0 resection rate. RESULTS Twenty-two patients enrolled in the study. Median follow-up during data cutoff was 23.9 months. There were no dose-limiting toxicities during the run-in phase. Nineteen patients (86.4%) underwent resection with R0 resection rate of 78.9% and with pCR rate of 26%. Most common treatment-related adverse events (TRAE) were cytopenias from chemoradiation. Aside from one grade ≥ 3 avelumab-related hypersensitivity, no grade ≥ 3 avelumab TRAEs were seen. Median RFS was not reached, and 1-year RFS and overall survival were 71% and 81%, respectively. The study was terminated before full planned accrual due to standard practice change based on the CheckMate 577 trial. CONCLUSIONS The addition of perioperative avelumab to nCRT was tolerable and demonstrated promising outcomes.
Collapse
Affiliation(s)
- Nataliya V Uboha
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Carbone Cancer Center, Madison, Wisconsin, USA
| | - Mustafa M Basree
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Jens C Eickhoff
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Dustin A Deming
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Carbone Cancer Center, Madison, Wisconsin, USA
| | - Kristina Matkowskyj
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - James Maloney
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
- Division of Cardiothoracic Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Daniel McCarthy
- Division of Cardiothoracic Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Malcolm DeCamp
- Division of Cardiothoracic Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Noelle LoConte
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Carbone Cancer Center, Madison, Wisconsin, USA
| | - Philip B Emmerich
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Sean Kraus
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Monica A Patel
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Carbone Cancer Center, Madison, Wisconsin, USA
| | - Jeremy D Kratz
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Carbone Cancer Center, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
- Center for Human Genomics and Precision Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Sam J Lubner
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Carbone Cancer Center, Madison, Wisconsin, USA
| | - Newton Hurst
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Michael F Bassetti
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
41
|
Song CW, Kim H, Kim MS, Park HJ, Paek SH, Terezakis S, Cho LC. Role of HIF-1α in the Responses of Tumors to Radiotherapy and Chemotherapy. Cancer Res Treat 2025; 57:1-10. [PMID: 38853541 PMCID: PMC11729307 DOI: 10.4143/crt.2024.255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024] Open
Abstract
Tumor microenvironment is intrinsically hypoxic with abundant hypoxia-inducible factors-1α (HIF-1α), a primary regulator of the cellular response to hypoxia and various stresses imposed on the tumor cells. HIF-1α increases radioresistance and chemoresistance by reducing DNA damage, increasing repair of DNA damage, enhancing glycolysis that increases antioxidant capacity of tumors cells, and promoting angiogenesis. In addition, HIF-1α markedly enhances drug efflux, leading to multidrug resistance. Radiotherapy and certain chemotherapy drugs evoke profound anti-tumor immunity by inducing immunologic cell death that release tumor-associated antigens together with numerous pro-immunological factors, leading to priming of cytotoxic CD8+ T cells and enhancing the cytotoxicity of macrophages and natural killer cells. Radiotherapy and chemotherapy of tumors significantly increase HIF-1α activity in tumor cells. Unfortunately, HIF-1α effectively promotes various immune suppressive pathways including secretion of immune suppressive cytokines, activation of myeloid-derived suppressor cells, activation of regulatory T cells, inhibition of T cells priming and activity, and upregulation of immune checkpoints. Consequently, the anti-tumor immunity elevated by radiotherapy and chemotherapy is counterbalanced or masked by the potent immune suppression promoted by HIF-1α. Effective inhibition of HIF-1α may significantly increase the efficacy of radiotherapy and chemotherapy by increasing radiosensitivity and chemosensitivity of tumor cells and also by upregulating anti-tumor immunity.
Collapse
Affiliation(s)
- Chang W Song
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Hyunkyung Kim
- Department of Radiation Oncology, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Mi-Sook Kim
- Department of Radiation Oncology, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Heon J Park
- Department of Microbiology, College of Medicine, Inha University, Incheon, Korea
| | - Sun-Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Korea
| | - Stephanie Terezakis
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - L Chinsoo Cho
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
42
|
Georgiou CJ, Brown MK, Cai Z, Alshafai L, Gao A, Rutka JT, Winnik MA, Reilly RM. Convection-enhanced delivery of [ 177Lu]Lu-labeled gold nanoparticles combined with anti-PD1 checkpoint immunotherapy improves the survival of immunocompetent C57BL/6J mice with orthotopic GL261 murine glioma tumors. Nucl Med Biol 2025; 140-141:108970. [PMID: 39571483 DOI: 10.1016/j.nucmedbio.2024.108970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 03/15/2025]
Abstract
INTRODUCTION Our objective was to study convection enhanced delivery (CED) of 177Lu-labeled metal chelating polymer (MCP) conjugated to gold nanoparticles ([177Lu]Lu-MCP-AuNP) alone or combined with anti-PD1 immune checkpoint inhibition (ICI) for improving the survival of immunocompetent C57BL/6J mice with orthotopic GL261 murine glioma tumors. METHODS C57BL/6J mice with GL261 tumors were treated with [177Lu]Lu-MCP-AuNP (0.8 or 2.7 MBq; 4 × 1011 AuNP) alone or combined with anti-PD1 antibodies (200 μg i.p. every 2 d × 3 doses). Control mice received normal saline, non-radioactive MCP-AuNP or anti-PD1 antibodies. Kaplan-Meier median survival was estimated. T-cell infiltration into the brain was probed by flow cytometry. Toxicity was assessed by monitoring body weight and cognitive function tests [Object Location Test (OLT) and Novel Object Recognition Test (NORT)] and T2-weighted MRI of the brain, overall health and ex vivo histopathological examination of the brain. RESULTS Treatment with [177Lu]Lu-MCP-AuNP (0.8 MBq) significantly increased median survival compared to MCP-AuNP (29 vs. 25 d; P = 0.007) or normal saline-treated mice (24 d; P < 0.001). Combining [177Lu]Lu-MCP-AuNP (0.8 MBq) with anti-PD1 antibodies increased median survival to 32 d (P < 0.0001 vs. normal saline). Increasing the mean amount of [177Lu]Lu-MCP-AuNP to 2.7 MBq and combining with anti-PD1 antibodies extended survival to at least 218 d in 5/9 mice. Increased CD8+ cytotoxic T-cells and decreased CD4+ helper T-cells were found in the brain vs. normal saline-treated mice. No weight loss (>20 %) was observed for treated or control mice. There was no change in cognitive function in mice treated with [177Lu]Lu-MCP-AuNP (0.8 MBq) alone or combined with anti-PD1 antibodies assessed by the OLT or NORT. T2-weighted MRI in mice treated with 2.7 MBq [177Lu]Lu-MCP-AuNP combined with anti-PD1 antibodies revealed edema, gliosis and ex vacuo dilatation of the ventricle proximal to the site of infusion. Histopathological examination of the brain revealed dilatation of the ventricle and gliosis proximal to the site of infusion but no radiation necrosis. MRI and histological analysis did not reveal tumor in the brain of these mice. Mice treated with 2.7 MBq [177Lu]Lu-MCP-AuNP combined with anti-PD1 antibodies did not demonstrate overall deleterious health effects. CONCLUSIONS We conclude that CED of [177Lu]Lu-MCP-AuNP combined with anti-PD1 checkpoint immunotherapy improved the survival of immunocompetent C67BL/6J mice with GL261 glioma tumors in the brain. Higher administered amounts of [177Lu]Lu-MCP-AuNP (2.7 MBq vs. 0.8 MBq) were most effective and yielded long-term survival. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE This study demonstrates that combining a locally-infused radiation nanomedicine, [177Lu]Lu-MCP-AuNP and anti-PD1 checkpoint immunotherapy improved the survival of mice with glioma tumors in the brain. In the future, this treatment may be useful to treat residual tumor at the surgical margins in patients with GBM to prevent local recurrence and improve survival.
Collapse
Affiliation(s)
| | - Madeline K Brown
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Laila Alshafai
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada; Joint Department of Medical Imaging, Division of Neuroradiology, Mount Sinai Hospital and University Health Network, Toronto, ON, Canada
| | - Andrew Gao
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
| | - James T Rutka
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada; Division of Neurosurgery, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada; Department of Medical Imaging, University of Toronto, Toronto, ON, Canada; Laboratory Medicine Program, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
43
|
Chekaoui A, Garofalo M, Gad B, Staniszewska M, Chiaro J, Pancer K, Gryciuk A, Cerullo V, Salmaso S, Caliceti P, Masny A, Wieczorek M, Pesonen S, Kuryk L. Cancer vaccines: an update on recent achievements and prospects for cancer therapy. Clin Exp Med 2024; 25:24. [PMID: 39720956 DOI: 10.1007/s10238-024-01541-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/17/2024] [Indexed: 12/26/2024]
Abstract
Decades of basic and translational research have led to a momentum shift in dissecting the relationship between immune cells and cancer. This culminated in the emergence of breakthrough immunotherapies that paved the way for oncologists to manage certain hard-to-treat cancers. The application of high-throughput techniques of genomics, transcriptomics, and proteomics was conclusive in making and expediting the manufacturing process of cancer vaccines. Using the latest research technologies has also enabled scientists to interpret complex and multiomics data of the tumour mutanome, thus identifying new tumour-specific antigens to design new generations of cancer vaccines with high specificity and long-term efficacy. Furthermore, combinatorial regimens of cancer vaccines with immune checkpoint inhibitors have offered new therapeutic approaches and demonstrated impressive efficacy in cancer patients over the last few years. In the present review, we summarize the current state of cancer vaccines, including their potential therapeutic effects and the limitations that hinder their effectiveness. We highlight the current efforts to mitigate these limitations and highlight ongoing clinical trials. Finally, a special focus will be given to the latest milestones expected to transform the landscape of cancer therapy and nurture hope among cancer patients.
Collapse
Affiliation(s)
- Arezki Chekaoui
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy.
| | - Beata Gad
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Monika Staniszewska
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Jacopo Chiaro
- Drug Research Program (DRP), ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE) University of Helsinki, Helsinki, Finland
- Translational Immunology Program (TRIMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, Helsinki, Finland
| | - Katarzyna Pancer
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Aleksander Gryciuk
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Vincenzo Cerullo
- Drug Research Program (DRP), ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE) University of Helsinki, Helsinki, Finland
- Translational Immunology Program (TRIMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, Helsinki, Finland
- Department of Molecular Medicine and Medical Biotechnology and CEINGE, University Federico II of Naples, Naples, Italy
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Aleksander Masny
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Magdalena Wieczorek
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | | | - Lukasz Kuryk
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland.
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy.
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland.
- Valo Therapeutics Oy, Helsinki, Finland.
| |
Collapse
|
44
|
Narra LR, Kumar R, Deek MP, Jabbour SK. Updates in Management of Unresectable Stage III Non Small Cell Lung Cancer: A Radiation Oncology Perspective. Cancers (Basel) 2024; 16:4233. [PMID: 39766132 PMCID: PMC11674665 DOI: 10.3390/cancers16244233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
Unresectable stage III non-small-cell lung cancer (NSCLC) remains a clinical challenge, due to the need for optimal local and systemic control. The management of unresectable Stage III NSCLC has evolved with advancements in radiation therapy (RT), systemic therapies, and immunotherapy. For patients with locally advanced NSCLC who are not surgical candidates, concurrent chemoradiotherapy (CRT) has modest survival outcomes, due to both local progression and distant metastasis. Efforts to enhance outcomes have led to dose-escalation trials, advances in modern RT techniques such as intensity-modulated RT (IMRT) and proton beam therapy (PBT), and the integration of adaptive RT to optimize target coverage while sparing organs at risk. Concurrent and consolidative immunotherapy, particularly with PD-L1 inhibitors, has shown promise, as evidenced by the PACIFIC trial, which demonstrated improved progression-free survival (PFS) and overall survival (OS) with durvalumab following CRT. Ongoing trials are now investigating novel immunotherapy combinations and targeted therapies in this setting, including dual checkpoint inhibition, DNA repair inhibitors, and molecularly targeted agents like osimertinib for EGFR-mutated NSCLC. Emerging biomarkers, such as circulating tumor DNA and radiomics, offer potential for personalizing treatment and predicting outcomes. Additionally, PBT and MR-guided adaptive RT have shown the potential to reduce toxicities while maintaining efficacy. Integrating these novel approaches may offer opportunities for optimizing treatment responses and minimizing adverse effects in this challenging patient population. Further investigation into patient stratification, biomarker-driven therapy, and refined therapeutic combinations is essential to improve long-term outcomes in unresectable Stage III NSCLC. This narrative review explores the current management strategies for unresectable Stage III NSCLC, from a radiation oncology perspective.
Collapse
Affiliation(s)
| | | | | | - Salma K. Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute, New Brunswick, NJ 08901, USA; (L.R.N.); (R.K.); (M.P.D.)
| |
Collapse
|
45
|
Xie Y, Liu Y, Lin M, Li Z, Shen Z, Yin S, Zheng Y, Zou Y, Zhang Y, Zhan Y, Fang Y, Ding Y. Targeting ATM enhances radiation sensitivity of colorectal cancer by potentiating radiation-induced cell death and antitumor immunity. J Adv Res 2024:S2090-1232(24)00601-5. [PMID: 39708961 DOI: 10.1016/j.jare.2024.12.023] [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: 06/01/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION The efficacy of radiotherapy in colorectal cancer (CRC) is often limited by radiation resistance. Ataxia telangiectasia mutated (ATM) is well known for its role in repairing double-strand DNA breaks within the DNA damage response (DDR) pathway. However, whether ATM mediates other mechanisms contributing to radiation resistance remains insufficiently investigated. OBJECTIVES This study investigates how targeting ATM enhances CRC radiation sensitivity and evaluates combination strategies to improve radiotherapy outcomes. METHODS Clinical specimens were analyzed to correlate ATM activation with radiotherapy response. Functional assays, including EdU, cell viability, clonogenic survival, and apoptosis assays, were used to assess the impact of ATM inhibition on radiation sensitivity. Mechanistic insights were gained through RNA-seq, RT-qPCR, western blotting, ELISA, immunofluorescence, flow cytometry, ChIP-qPCR, and co-immunoprecipitation. In vivo efficacy was evaluated using subcutaneous tumor models in nude, BALB/c, and C57BL/6J mice. RESULTS High ATM phosphorylation levels correlated with poor radiotherapy response in CRC patients. ATM inhibition enhanced radiation sensitivity in both in vitro and in vivo models. Mechanistically, ATM inhibition increased radiation-induced ROS accumulation and mitochondrial damage, leading to the release of mitochondrial DNA (mtDNA) into the cytosol and activation of the STING-type I interferon pathway. This enhanced CD8+ T cell infiltration and boosted antitumor immunity. Additionally, ATM inhibition partially alleviated the radiation-induced upregulation of PD-L1, likely through the ATM/NEMO/NF-κB pathway. Notably, triple therapy combining radiotherapy, an ATM inhibitor, and anti-PD-L1 achieved superior tumor control and remission in mouse models, including large, treatment-resistant tumors. CONCLUSION Targeting ATM enhances radiation-induced tumor cell death and boosts antitumor immune responses, offering a promising strategy to overcome CRC radiation resistance. The synergy of radiotherapy, ATM inhibitior, and immune checkpoint blockade highlights a novel therapeutic approach for CRC management.
Collapse
Affiliation(s)
- Yuwen Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China; Department of Radiation Oncology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan, China
| | - Yang Liu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Mingdao Lin
- Department of Anorectal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan, China
| | - Zhenkang Li
- Department of Surgery, Zhujiang Hospital, Southern Medical University Guangzhou 510280, Guangdong, China
| | - Zhiyong Shen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shengqi Yin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yilin Zheng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yishu Zou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yaowei Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yizhi Zhan
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China.
| | - Yuan Fang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China.
| | - Yi Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China.
| |
Collapse
|
46
|
Zhang Y, Zeng Y, Yin Y, Zhang W, Li T, Jiang T, Zheng X, Yu Z, Cai X, Zhang Q. The role of radiotherapy in extensive-stage small cell lung cancer: insights from treatment failure patterns in the era of immunotherapy. BMC Cancer 2024; 24:1534. [PMID: 39695991 DOI: 10.1186/s12885-024-13297-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The therapeutic advantage of radiotherapy (RT) as an adjunct to first-line immunotherapy and chemotherapy in patients with extensive-stage small cell lung cancer (ES-SCLC) remains unclear. This study aimed to elucidate the value of RT based on the first failure pattern of ES-SCLC. METHODS In this study, we retrospectively analyzed ES-SCLC patients treated with first-line chemotherapy and immune checkpoint inhibitors (ICIs) at Shanghai Chest Hospital from August 2018 to October 2023. Our study recorded the first failure pattern in ES-SCLC, analyzed the main sites of disease progression, explored factors that may affect prognosis and estimated the value of RT in extending patient survival in the immunotherapy era. Key endpoints included the first failure pattern, progression-free survival (PFS) and overall survival (OS). RESULTS Among 344 patients, 70 (20%) had local failure, 105 (31%) had distant failure, 69 (20%) experienced both types of failure, and 100 (29%) showed no disease progression. Disease progression occurred in 244 patients (71%). They were divided into two groups: 183 without pre-progression RT and 61 with pre-progression RT. In the non-pre-progression RT group, 55 patients (30%) had local failure, 72 patients (39%) had distant failure, and 56 patients (31%) had both. In the pre-progression RT group, 15 patients (25%) had local failure, 33 patients (54%) had distant failure, and 13 patients (21%) had both. Univariate and multivariate analyses identified RT as an independent prognostic factor for improved OS (P < 0.05). Subgroup analysis further confirmed these findings. Pre-progression RT was associated with superior PFS (P < 0.05). The median overall survival (mOS) was 20.1 months (95% confidence interval [CI]: 15.5-24.7 months) in the pre-progression RT group, compared to 13.4 months (95% CI: 13.4-19.2 months) in the non-pre-progression RT group. CONCLUSION RT improved OS in these patients, and pre-progression RT improved PFS further. Pre-progression RT shows a trend toward OS benefit.
Collapse
Affiliation(s)
- Ya Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, People's Republic of China
| | - Ya Zeng
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, People's Republic of China
| | - Yipengchen Yin
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, People's Republic of China
| | - Wenqing Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, People's Republic of China
| | - Tianyu Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China
| | - Tiaoyan Jiang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, People's Republic of China
| | - Xiaojun Zheng
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, People's Republic of China
| | - Zhongdan Yu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, People's Republic of China
| | - Xuwei Cai
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, People's Republic of China.
| | - Qin Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, People's Republic of China.
| |
Collapse
|
47
|
Hong S, Park J, Oh Y, Cho H, Kim K. Nanotechnology-Based Strategies for Safe and Effective Immunotherapy. Molecules 2024; 29:5855. [PMID: 39769944 PMCID: PMC11676242 DOI: 10.3390/molecules29245855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer immunotherapy using immune checkpoint blockades has emerged as a promising therapeutic approach. However, immunotherapy faces challenges such as low response rates in solid tumors, necessitating strategies to remodel the immune-suppressive tumor microenvironment (TME) into an immune-activated state. One of the primary approaches to achieve this transformation is through the induction of immunogenic cell death (ICD). Herein, we discussed strategies to maximize ICD induction using nanoparticles. In particular, this review highlighted various studies integrating chemotherapy, radiation therapy (RT), photodynamic therapy (PDT), and photothermal therapy (PTT) with nanoparticle-based immunotherapy. The research covered in this review aims to provide valuable insights for future studies on nanoparticle-assisted immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; (S.H.); (J.P.); (Y.O.); (H.C.)
| |
Collapse
|
48
|
Cintoni M, Palombaro M, Raoul P, Chiloiro G, Romano A, Meldolesi E, De Giacomo F, Leonardi E, Egidi G, Grassi F, Pulcini G, Rinninella E, Capristo E, Gasbarrini A, Gambacorta MA, Mele MC. Assessing Quality of Life with the Novel QLQ-CAX24 Questionnaire and Body Composition Parameters in Rectal Cancer Patients: A Single-Center Prospective Study. Nutrients 2024; 16:4277. [PMID: 39770899 PMCID: PMC11678168 DOI: 10.3390/nu16244277] [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: 11/12/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Patients with rectal cancer (RC) are at risk of developing cancer-related cachexia, a complex metabolic syndrome that can negatively impact quality of life (QoL), treatment tolerance, and clinical response. OBJECTIVES The aim of the study was to explore the possible associations of the novel European Organization for Research and Treatment of Cancer QoL Questionnaire-Cancer Cachexia (EORTC QLQ-CAX24) scores with body composition parameters and physical performance in patients with locally advanced RC (LARC). METHODS This prospective observational study involved RC patients evaluated at the dedicated outpatient clinic of Clinical Nutrition at the Fondazione Policlinico Agostino Gemelli IRCCS. Patients with a confirmed diagnosis of LARC were enrolled between January and December 2023. The body composition parameters were measured using the preoperative computed tomography scan at the level of the third lumbar vertebra as well as using bioimpedance analysis before and after the radiotherapy treatment. QoL was measured by the EORTC QLQ-C30 and EORTC QLQ-CAX24 questionnaires. RESULTS A total of 56 RC patients were enrolled. Significant associations (p < 0.05) were found between EORTC QLQ-CAX24 values and the presence of cachexia, body composition, handgrip strength, and malnutrition diagnosis. Muscle mass was significatively also associated with EORTC QLQ-CAX24 results, suggesting a link between subjective perception of QoL and objectively measured body composition. CONCLUSIONS The EORTC CAX24 questionnaire can be an effective tool for monitoring changes in cachexia status during radiotherapy, enabling early detection of cachexia-related complications and timely intervention.
Collapse
Affiliation(s)
- Marco Cintoni
- UOC di Nutrizione Clinica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (M.C.); (M.P.); (E.L.); (G.E.); (F.G.); (G.P.); (E.R.); (M.C.M.)
- Centro di Ricerca e Formazione in Nutrizione Umana, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.C.)
| | - Marta Palombaro
- UOC di Nutrizione Clinica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (M.C.); (M.P.); (E.L.); (G.E.); (F.G.); (G.P.); (E.R.); (M.C.M.)
| | - Pauline Raoul
- UOC di Nutrizione Clinica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (M.C.); (M.P.); (E.L.); (G.E.); (F.G.); (G.P.); (E.R.); (M.C.M.)
| | - Giuditta Chiloiro
- UOC Servizio di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (G.C.); (A.R.); (E.M.); (F.D.G.); (M.A.G.)
| | - Angela Romano
- UOC Servizio di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (G.C.); (A.R.); (E.M.); (F.D.G.); (M.A.G.)
| | - Elisa Meldolesi
- UOC Servizio di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (G.C.); (A.R.); (E.M.); (F.D.G.); (M.A.G.)
| | - Flavia De Giacomo
- UOC Servizio di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (G.C.); (A.R.); (E.M.); (F.D.G.); (M.A.G.)
| | - Elena Leonardi
- UOC di Nutrizione Clinica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (M.C.); (M.P.); (E.L.); (G.E.); (F.G.); (G.P.); (E.R.); (M.C.M.)
| | - Gabriele Egidi
- UOC di Nutrizione Clinica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (M.C.); (M.P.); (E.L.); (G.E.); (F.G.); (G.P.); (E.R.); (M.C.M.)
| | - Futura Grassi
- UOC di Nutrizione Clinica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (M.C.); (M.P.); (E.L.); (G.E.); (F.G.); (G.P.); (E.R.); (M.C.M.)
| | - Gabriele Pulcini
- UOC di Nutrizione Clinica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (M.C.); (M.P.); (E.L.); (G.E.); (F.G.); (G.P.); (E.R.); (M.C.M.)
| | - Emanuele Rinninella
- UOC di Nutrizione Clinica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (M.C.); (M.P.); (E.L.); (G.E.); (F.G.); (G.P.); (E.R.); (M.C.M.)
- Centro di Ricerca e Formazione in Nutrizione Umana, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.C.)
| | - Esmeralda Capristo
- Centro di Ricerca e Formazione in Nutrizione Umana, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.C.)
- UOS Medicina della Grande Obesità, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Centro di Ricerca e Formazione in Nutrizione Umana, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.C.)
- UOC Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Maria Antonietta Gambacorta
- UOC Servizio di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (G.C.); (A.R.); (E.M.); (F.D.G.); (M.A.G.)
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Maria Cristina Mele
- UOC di Nutrizione Clinica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (M.C.); (M.P.); (E.L.); (G.E.); (F.G.); (G.P.); (E.R.); (M.C.M.)
- Centro di Ricerca e Formazione in Nutrizione Umana, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.C.)
| |
Collapse
|
49
|
Ganina A, Askarov M, Kozina L, Karimova M, Shayakhmetov Y, Mukhamedzhanova P, Brimova A, Berikbol D, Chuvakova E, Zaripova L, Baigenzhin A. Prospects for Treatment of Lung Cancer Using Activated Lymphocytes Combined with Other Anti-Cancer Modalities. Adv Respir Med 2024; 92:504-525. [PMID: 39727496 PMCID: PMC11673795 DOI: 10.3390/arm92060045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
This review explores the significance and prospects of using diverse T-cell variants in the context of combined therapy for lung cancer treatment. Recently, there has been an increase in research focused on understanding the critical role of tumor-specific T lymphocytes and the potential benefits of autologous T-cell-based treatments for individuals with lung cancer. One promising approach involves intravenous administration of ex vivo-activated autologous lymphocytes to improve the immune status of patients with cancer. Investigations are also exploring the factors that influence the success of T-cell therapy and the methods used to stimulate them. Achieving a comprehensive understanding of the characteristics of activated lymphocytes and deciphering the mechanisms underlying their activation of innate anti-tumor immunity will pave the way for numerous clinical trials and the development of innovative strategies for cancer therapy like combined immunotherapy and radiation therapy.
Collapse
Affiliation(s)
- Anastasia Ganina
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Manarbek Askarov
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Larissa Kozina
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Madina Karimova
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Yerzhan Shayakhmetov
- International Oncological Tomotherapy Center “YMIT”, Astana 010009, Kazakhstan; (Y.S.); (P.M.); (A.B.); (D.B.)
| | - Perizat Mukhamedzhanova
- International Oncological Tomotherapy Center “YMIT”, Astana 010009, Kazakhstan; (Y.S.); (P.M.); (A.B.); (D.B.)
| | - Aigul Brimova
- International Oncological Tomotherapy Center “YMIT”, Astana 010009, Kazakhstan; (Y.S.); (P.M.); (A.B.); (D.B.)
| | - Daulet Berikbol
- International Oncological Tomotherapy Center “YMIT”, Astana 010009, Kazakhstan; (Y.S.); (P.M.); (A.B.); (D.B.)
| | - Elmira Chuvakova
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Lina Zaripova
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| | - Abay Baigenzhin
- JSC National Scientific Medical Center, Astana 010009, Kazakhstan; (M.A.); (L.K.); (M.K.); (E.C.); (L.Z.); (A.B.)
| |
Collapse
|
50
|
Xue H, Chen Y, Zhou Y. Radioimmunotherapy: a game-changer for advanced non-small cell lung cancer. Front Immunol 2024; 15:1522508. [PMID: 39712010 PMCID: PMC11659256 DOI: 10.3389/fimmu.2024.1522508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Lung cancer, particularly non-small cell lung cancer (NSCLC), remains a leading cause of cancer-related deaths, with conventional treatments offering limited effectiveness in advanced stages, due to distant metastases and treatment resistance. Recent advancements in immunotherapy, specifically immune checkpoint inhibitors (ICIs), have shown promise, but their efficacy as standalone therapies are often insufficient. This has led to increased interest in combining ICIs with radiotherapy, known as radioimmunotherapy (iRT), to enhance treatment outcomes. This review explores the mechanisms that underlie the synergy between radiotherapy and immunotherapy. Radiotherapy can induce the "abscopal effect", eliciting systemic immune responses that reduce tumor burdens outside the treated area. It also increases the expression of major histocompatibility complex class I (MHC-I) on tumor cells, improving immune recognition. Furthermore, radiotherapy can modify the tumor microenvironment by inducing metabolic reprogramming to bolster anti-tumor immunity. We discuss strategies for optimizing iRT, including considerations of radiation doses, fractionation schedules, and treatment site selection, which significantly influence immune responses by enhancing MHC-I expression or promoting T-cell infiltration. Clinical evidence supports the efficacy of iRT in NSCLC and other cancers, though challenges in standardizing treatment protocols and managing side effects persist. Overall, radioimmunotherapy presents a promising approach to improving NSCLC treatment outcomes. Ongoing research into its mechanisms and the refinement of treatment may reshape clinical practice, offering more effective and personalized options for patients with advanced lung cancer. Further studies are essential to validate these findings and optimize therapeutic protocols.
Collapse
Affiliation(s)
- Huichan Xue
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Yunshang Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Yun Zhou
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|