1
|
Gao Y, Yu B, Li L, Zhang J, Zhao T, Feng X, Hirayama R, Di C, Zhang Y, Ye Y, Li Y, Li Q, Jin X. mtDNA/RNA boosts radiation-induced abscopal effect via M1 macrophage polarization-promoted IFN-β-dependent inflammatory response. Int Immunopharmacol 2025; 155:114673. [PMID: 40245773 DOI: 10.1016/j.intimp.2025.114673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/28/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
The radiation-induced abscopal effect (RIAE) can suppress distal metastatic lesions and elicit a systemic anti-tumor response; however, the underlying mechanisms remain to be fully elucidated. Current research has shown that autophagy promotes the production of IFN-β by regulating mitochondrial DNA (mtDNA), thereby contributing to the modulation of RIAE. Nevertheless, the downstream pathways through which IFN-β influences RIAE require further investigation. In this study, we observed accumulation of an abundance of mtDNA in the cytosol of mammary tumor cells following RT, along with the presence of mitochondrial RNA (mtRNA). These molecules activated the cGAS-STING and RIG-I-MAVS signaling pathways, respectively, thereby synergistically promoting the production of IFN-β and secretion into the extracellular matrix. Subsequently, IFN-β facilitated the polarization of macrophages in distant non-irradiated tumor microenvironment towards the M1 phenotype through activating STAT1. Furthermore, our findings indicate that high linear energy transfer (LET) carbon ions are significantly more effective in inducing the production of IFN-β and promoting macrophage polarization compared to low-LET X-rays. Thus, our findings provide insights into the intricate mechanisms by which mtDNA/RNA and IFN-β mediate RIAE, suggesting that IFN-β could be a promising target for provoking RT immunogenicity in patients with breast cancer and high-LET radiation might effectively elicit RIAE.
Collapse
Affiliation(s)
- Yuting Gao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; School of Life Sciences, Northwest Normal University, Gansu Province, Lanzhou 730070, China
| | - Boyi Yu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Linjing Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiahao Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Ting Zhao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | - Xianglong Feng
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ryoichi Hirayama
- National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Cuixia Di
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanshan Zhang
- Gansu Wuwei Tumor Hospital, Wuwei 733000, Gansu Province, China
| | - Yancheng Ye
- Gansu Wuwei Tumor Hospital, Wuwei 733000, Gansu Province, China
| | - Yuan Li
- School of Life Sciences, Northwest Normal University, Gansu Province, Lanzhou 730070, China.
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
2
|
Yanagie H, Maruyama S, Oyama K, Ono Y, Kuroyama S, Nonaka Y, Hatae R, Furuya Y, Suzuki M, Masunaga SI, Sakurai Y, Kondo N, Tanaka H, Maruhashi A, Ono K, Sugihara T, Takahashi H. First Experiences of Pilot Clinical Studies on Boron Neutron Capture Therapy for Recurrent Gastrointestinal Cancers Using an Intravenous Injection of 10BPA. In Vivo 2025; 39:1470-1491. [PMID: 40294997 PMCID: PMC12041971 DOI: 10.21873/invivo.13948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND/AIM Boron neutron capture therapy (BNCT) is a novel treatment that induces targeted tumor cell damage through the selective accumulation of 10B compounds in cancer cells followed by the production of alpha and lithium particles using thermal neutron irradiation. Despite its potential, clinical applications of BNCT for recurrent gastrointestinal cancers remain limited. This study presents the first pilot clinical evaluation of BNCT using intravenous boronophenylalanine (10BPA) for such cancers. CASE REPORTS Four patients with recurrent gastrointestinal cancers were enrolled in this phase I-II clinical study. All had tumors refractory to standard treatments, including surgery, chemotherapy, and radiotherapy. BNCT was performed using thermal neutron irradiation at Kyoto University Research Reactor. 10BPA was administered intravenously at 400 mg/kg, and no severe adverse effects were observed. Tumor responses varied, with one patient achieving partial response and three demonstrating stable disease at three months post-treatment. Notably, BNCT alleviated cancer-related symptoms, such as pain and nerve compression, improving patients' quality of life. Dosimetric evaluations confirmed effective tumor doses with acceptable exposure to surrounding normal tissues. CONCLUSION BNCT is a promising modality for recurrent gastrointestinal cancers, offering symptom relief and potential antitumor effects. Its safety and feasibility were demonstrated in this study. Future research should explore fractionated BNCT and combination therapies with immunotherapy or targeted agents to enhance efficacy further.
Collapse
Affiliation(s)
- Hironobu Yanagie
- Department of Surgery, Shin-Yamate Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan;
- Department of Gastrointestinal Surgery, Keiaikai Kojin Hospital, Iwate, Japan
- Institute of Engineering Innovation, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
- Cooperative Unit of Medicine & Engineering, The University of Tokyo Hospital, Tokyo, Japan
- Niigata University of Pharmacy and Applied Life Sciences, Niigata, Japan
| | - Syoji Maruyama
- Department of Surgery, Shin-Yamate Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Kazuyuki Oyama
- Department of Radiology, Shin-Yamate Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Yasuo Ono
- Department of Surgical Oncology, Kameda Medical Center, Chiba, Japan
- Department of Surgery, Fuchinobe General Hospital, Kanagawa, Japan
| | | | - Yasumasa Nonaka
- Department of Surgery, Keiaikai Houyo Hospital, Iwate, Japan
| | - Ryo Hatae
- Department of Surgery, Fuchinobe General Hospital, Kanagawa, Japan
| | - Yoshitaka Furuya
- Department of Surgery, Sodegaura Satsukidai Hospital, Chiba, Japan
| | - Minoru Suzuki
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka, Japan
| | - Shin-Ichiro Masunaga
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka, Japan
- BNCT Research Center, Organization for Research Promotion, Osaka Metropolitan University, Osaka, Japan
- Kinshukai, Hanwadaini Senhoku Hospital, Osaka, Japan
| | - Yoshinori Sakurai
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka, Japan
| | - Natsuko Kondo
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka, Japan
| | - Hiroki Tanaka
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka, Japan
| | - Akira Maruhashi
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka, Japan
| | - Koji Ono
- Kansai BNCT Medical Center, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Takumichi Sugihara
- Department of Radiology, Shin-Yamate Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Hiroyuki Takahashi
- Institute of Engineering Innovation, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
- Cooperative Unit of Medicine & Engineering, The University of Tokyo Hospital, Tokyo, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
3
|
Ma L, Li Y, Sakamoto Y, Xie L, Suzuki S, Yoshida Y, Sui L, Guo G, Wen J, Ren W, Kakimi K, Osada K, Takahashi A, Shimokawa T. Optimal radiation dose to induce an abscopal effect by combining carbon-ion radiotherapy and anti-CTLA4 antibody. Neoplasia 2025; 60:101099. [PMID: 39674115 PMCID: PMC11699741 DOI: 10.1016/j.neo.2024.101099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND AND PURPOSE Although carbon-ion radiotherapy (CIRT) has led to good outcomes, controlling metastasis is still crucial for improving overall survival. This study aimed to evaluate the effectiveness of by two combinations, one of CIRT and anti-CTLA4 antibody, the other of CIRT and anti-PD-1 antibody, applied at different radiation doses for distal tumour and metastasis suppression. MATERIALS AND METHODS Murine cancer cells (colon carcinoma Colon-26 cells for experiments and osteosarcoma LM8 cells for verification) were grafted into both sides of the hind legs of syngeneic mice. Right-side tumours were irradiated with 3 Gy or 10 Gy CIRT while the left-side tumours were not irradiated, followed by the administration of the anti-CTLA4 antibody or anti-PD-1 antibody. The diameter of the tumours in both legs was measured 3 times per week after irradiation. The number of pulmonary metastases was evaluated within 3 weeks after irradiation. RESULTS Compared with the control group, the high-dose group showed promising anti-cancer benefits in terms of both irradiated tumours and lung metastasis, but neither 10 Gy CIRT combined with the anti-CTLA4 antibody nor 10 Gy CIRT combined with the anti-PD-1 antibody suppressed the growth of distant unirradiated tumours. In the low-dose group, the effect on primary tumour control was slightly weaker than that in the high-dose treatment group, but significant suppressive effects on both distant unirradiated tumours and metastases were observed following 3 Gy CIRT combined with anti-CTLA4 antibody treatment. Specifically, the volume of distant unirradiated tumours decreased by 40 % compared with that of the control group, and no lung metastasis was observed. CONCLUSION Our findings suggest that there is an optimal dose range for the abscopal effect generated with the CIRT combined with anti-CTLA4 antibody, and it highlights a new opportunity for increased induction efficiency of the abscopal effect of combination therapy.
Collapse
Affiliation(s)
- Liqiu Ma
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan; Gunma University Heavy Ion Medical Center, Gunma 371-8511, Japan; Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China.
| | - Yang Li
- Gunma University Heavy Ion Medical Center, Gunma 371-8511, Japan
| | - Yoshimitsu Sakamoto
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | - Lin Xie
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | - Saaya Suzuki
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | - Yukari Yoshida
- Gunma University Heavy Ion Medical Center, Gunma 371-8511, Japan
| | - Li Sui
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China
| | - Gang Guo
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China
| | - Jialing Wen
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China
| | - Wangcai Ren
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China
| | - Kazuhiro Kakimi
- Department of Immunology, Kindai University Faculty of Medicine, Osaka 589-0014, Japan
| | - Kensuke Osada
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | | | - Takashi Shimokawa
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan.
| |
Collapse
|
4
|
Ren W, Wen J, Guo G, Gu W, Zhang S, Liu C, Osada K, Shimokawa T, Wang Q, Wang Y, Tu X, Li C, Sui L, Ma L. Physical parameters and biological factors affect the abscopal effect of combining radiotherapy with immunotherapy: an update on preclinical works. Front Public Health 2025; 12:1517147. [PMID: 39949344 PMCID: PMC11822297 DOI: 10.3389/fpubh.2024.1517147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/20/2024] [Indexed: 02/16/2025] Open
Abstract
In the process of radiotherapy for cancer patients, there is an extremely low probability phenomenon that the distal tumor/metastasis away from the irradiation field undergoes regression after localized radiation therapy, which is called the abscopal effect. Enhancing the incidence of this phenomenon possesses profound significance for the investigation of metastatic cancer treatment. Currently, the underlying mechanisms of the abscopal effect remain unclear. Radiation-induced immunogenic cell death is considered one of the potential mechanisms for the abscopal effect. From this perspective, we explored how physical parameters and biological factors influence this process. Differences between patients with respect to physical factors and intrinsic biological factors that activate the immune response (acquired factors) may affect the induction of the abscopal effect.
Collapse
Affiliation(s)
- Wangcai Ren
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing, China
| | - Jialing Wen
- National Innovation Center of Radiation Application, Beijing, China
| | - Gang Guo
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing, China
- National Innovation Center of Radiation Application, Beijing, China
| | - Wenchao Gu
- Department of Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shenke Zhang
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Chang Liu
- Department of Accelerator and Medical Physics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Kensuke Osada
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Takashi Shimokawa
- Department of Accelerator and Medical Physics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Qiaojuan Wang
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing, China
- National Innovation Center of Radiation Application, Beijing, China
| | - Yue Wang
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing, China
- National Innovation Center of Radiation Application, Beijing, China
| | - Xuanzhang Tu
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing, China
| | - Chen Li
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Li Sui
- National Innovation Center of Radiation Application, Beijing, China
| | - Liqiu Ma
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing, China
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| |
Collapse
|
5
|
Romero Fernandez J, Cordoba Largo S, Benlloch Rodriguez R, Gil Haro B. The Effects of Gynecological Tumor Irradiation on the Immune System. Cancers (Basel) 2024; 16:2804. [PMID: 39199577 PMCID: PMC11352652 DOI: 10.3390/cancers16162804] [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: 07/04/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Radiobiology has evolved from a mechanistic model based on DNA damage and response factors into a more complex model that includes effects on the immune system and the tumor microenvironment (TME). Irradiation has an immunomodulatory effect that can manifest as increased anti-tumor immunity or immunosuppression. Irradiation promotes an inflammatory microenvironment through the release of pro-inflammatory cytokines and endothelial damage, which recruit immune system cells to the irradiated area. Radiation-induced immunogenic cell death (ICD), characterized by the release of damage-associated molecular patterns (DAMPs) and tumor antigens, triggers an anti-tumor immune response of both innate and adaptive immunity. Anti-tumor immunity can manifest at a distance from the irradiated area, a phenomenon known as the abscopal effect (AE), which involves dendritic cells and CD8+ T cells. Irradiation also produces an immunosuppressive effect mediated by tumor-associated macrophages (TAMs) and regulatory T lymphocytes (Tregs), which counterbalances the immunostimulatory effect. In this work, we review the mechanisms involved in the radiation-induced immune response, which support the combined treatment of RT and immunotherapy, focusing, where possible, on gynecologic cancer.
Collapse
Affiliation(s)
- Jesus Romero Fernandez
- Radiation Oncology Department, Hospital Universitario Puerta de Hierro, C. Joaquín Rodrigo 1, 28222 Majadahonda, Spain; (S.C.L.); (R.B.R.); (B.G.H.)
| | | | | | | |
Collapse
|
6
|
Zheng Z, Yang T, Li Y, Qu P, Shao Z, Wang Y, Chang W, Umar SM, Wang J, Ding N, Wang W. A future directions of renal cell carcinoma treatment: combination of immune checkpoint inhibition and carbon ion radiotherapy. Front Immunol 2024; 15:1428584. [PMID: 39091498 PMCID: PMC11291258 DOI: 10.3389/fimmu.2024.1428584] [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: 05/06/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Renal cell carcinoma (RCC) is considered radio- and chemo-resistant. Immune checkpoint inhibitors (ICIs) have demonstrated significant clinical efficacy in advanced RCC. However, the overall response rate of RCC to monotherapy remains limited. Given its immunomodulatory effects, a combination of radiotherapy (RT) with immunotherapy is increasingly used for cancer treatment. Heavy ion radiotherapy, specifically the carbon ion radiotherapy (CIRT), represents an innovative approach to cancer treatment, offering superior physical and biological effectiveness compared to conventional photon radiotherapy and exhibiting obvious advantages in cancer treatment. The combination of CIRT and immunotherapy showed robust effectiveness in preclinical studies of various tumors, thus holds promise for overcoming radiation resistance of RCC and enhancing therapeutic outcomes. Here, we provide a comprehensive review on the biophysical effects of CIRT, the efficacy of combination treatment and the underlying mechanisms involved in, as well as its therapeutic potential specifically within RCC.
Collapse
Affiliation(s)
- Zhouhang Zheng
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Tianci Yang
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Yixuan Li
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Pei Qu
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Zhiang Shao
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Wang
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Wei Chang
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Shahzad Muhammad Umar
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Jufang Wang
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Nan Ding
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Wei Wang
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
7
|
Salomon N, Helm A, Selmi A, Fournier C, Diken M, Schrörs B, Scholz M, Kreiter S, Durante M, Vascotto F. Carbon Ion and Photon Radiation Therapy Show Enhanced Antitumoral Therapeutic Efficacy With Neoantigen RNA-LPX Vaccines in Preclinical Colon Carcinoma Models. Int J Radiat Oncol Biol Phys 2024; 119:936-945. [PMID: 38163521 DOI: 10.1016/j.ijrobp.2023.12.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/07/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE Personalized liposome-formulated mRNA vaccines (RNA-LPX) are a powerful new tool in cancer immunotherapy. In preclinical tumor models, RNA-LPX vaccines are known to achieve potent results when combined with conventional X-ray radiation therapy (XRT). Densely ionizing radiation used in carbon ion radiation therapy (CIRT) may induce distinct effects in combination with immunotherapy compared with sparsely ionizing X-rays. METHODS AND MATERIALS Within this study, we investigate the potential of CIRT and isoeffective doses of XRT to mediate tumor growth inhibition and survival in murine colon adenocarcinoma models in conjunction with neoantigen (neoAg)-specific RNA-LPX vaccines encoding both major histocompatibility complex (MHC) class I- and class II-restricted tumor-specific neoantigens. We characterize tumor immune infiltrates and antigen-specific T cell responses by flow cytometry and interferon-γ enzyme-linked immunosorbent spot (ELISpot) analyses, respectively. RESULTS NeoAg RNA-LPX vaccines significantly potentiate radiation therapy-mediated tumor growth inhibition. CIRT and XRT alone marginally prime neoAg-specific T cell responses detected in the tumors but not in the blood or spleens of mice. Infiltration and cytotoxicity of neoAg-specific T cells is strongly driven by RNA-LPX vaccines and is accompanied by reduced expression of the inhibitory markers PD-1 and Tim-3 on these cells. The neoAg RNA-LPX vaccine shows similar overall therapeutic efficacy in combination with both CIRT and XRT, even if the physical radiation dose is lower for carbon ions than for X-rays. CONCLUSIONS We hence conclude that the combination of CIRT and neoAg RNA-LPX vaccines is a promising strategy for the treatment of radioresistant tumors.
Collapse
Affiliation(s)
- Nadja Salomon
- TRON gGmbH, Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Alexander Helm
- GSI Helmholtzzentrum for Heavy Ion Research GmbH, Darmstadt, Germany
| | - Abderaouf Selmi
- TRON gGmbH, Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudia Fournier
- GSI Helmholtzzentrum for Heavy Ion Research GmbH, Darmstadt, Germany
| | - Mustafa Diken
- TRON gGmbH, Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Barbara Schrörs
- TRON gGmbH, Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael Scholz
- GSI Helmholtzzentrum for Heavy Ion Research GmbH, Darmstadt, Germany
| | - Sebastian Kreiter
- TRON gGmbH, Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Marco Durante
- GSI Helmholtzzentrum for Heavy Ion Research GmbH, Darmstadt, Germany; Technical University Darmstadt, Institute of Condensed Matter Physics, Darmstadt, Germany; University Federico II, Department of Physics "Ettore Pancini", Naples, Italy
| | - Fulvia Vascotto
- TRON gGmbH, Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
8
|
Yu B, Gao Y, Li J, Gao F, Zhang J, Li L, Feng X, Zuo D, Jin X, Chen W, Li Q. Killing two birds with one stone: Abscopal effect mechanism and its application prospect in radiotherapy. Crit Rev Oncol Hematol 2024; 196:104325. [PMID: 38462151 DOI: 10.1016/j.critrevonc.2024.104325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/07/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024] Open
Abstract
Abscopal effects are characterized by the emergence of neoplasms in regions unrelated to the primary radiation therapy site, displaying a gradual attenuation or regression throughout the progression of radiation therapy, which have been of interest to scientists since Mole's proposal in 1953. The incidence of abscopal effects in radiation therapy is intricately linked to the immune system, with both innate and adaptive immune responses playing crucial roles. Biological factors impacting abscopal effects ultimately exert their influence on the intricate workings of the immune system. Although abscopal effects are rarely observed in clinical cases, the underlying mechanism remains uncertain. This article examines the biological and physical factors influencing abscopal effects of radiotherapy. Through a review of preclinical and clinical studies, this article aims to offer a comprehensive understanding of abscopal effects and proposes new avenues for future research in this field. The findings presented in this article serve as a valuable reference for researchers seeking to explore this topic in greater depth.
Collapse
Affiliation(s)
- Boyi Yu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuting Gao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; College of Life Sciences, Northwest Normal University, Gansu Province, Lanzhou 730070, China
| | - Jiaxin Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feifei Gao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiahao Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; School of Public Health, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Linjing Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianglong Feng
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dashan Zuo
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
9
|
Meng K, Lu H. Clinical application of high-LET radiotherapy combined with immunotherapy in malignant tumors. PRECISION RADIATION ONCOLOGY 2024; 8:42-46. [PMID: 40336565 PMCID: PMC11935250 DOI: 10.1002/pro6.1225] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 02/22/2024] [Indexed: 05/09/2025] Open
Abstract
The superior physical and biological properties of high linear energy transfer (LET), as opposed to traditional low-LET rays, underscore the advantage of proton therapy (PRT) and carbon ion radiotherapy (CIRT) are better than traditional photon radiotherapy (XRT). With the advancements in science, an increasing number of hospitals have introduced new technologies. However, radiotherapy is primarily used for local treatment, which means that if the tumor has metastasized to distant sites, it is often necessary to combine it with systemic therapies such as immunotherapy. In recent years, the combination of high-LET radiotherapy and immunotherapy has emerged as a promising treatment option in oncology and many studies have confirmed its efficacy for both local and distant metastases. In this review, we summarize the effects of PRT and CIRT on the immune system in detail, followed by an introduction to preclinical and clinical studies of PRT and CIRT in combination with immune checkpoint inhibitor (ICIs) therapy. We also briefly introduce some preclinical studies on CIRT in combination with dendritic cells (DCs) and Treg inhibitor therapies.
Collapse
Affiliation(s)
| | - Haijun Lu
- Department of RadiotherapyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| |
Collapse
|
10
|
Liu R, Geng Y, Luo H, Zhang Q, Yang Z, Li S, Sun S, Liu Z, Dong M, Du T, Che T, Wang X. Carbon ion irradiation combined with PD-1 inhibitor trigger abscopal effect in Lewis lung cancer via a threshold dose. J Cancer 2024; 15:2245-2259. [PMID: 38495488 PMCID: PMC10937268 DOI: 10.7150/jca.91559] [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: 10/26/2023] [Accepted: 01/15/2024] [Indexed: 03/19/2024] Open
Abstract
Background and goal: Carbon ion beam is radio-biologically more efficient than photons and is beneficial for treating radio-resistant tumors. Several animal experiments with tumor-bearing suggest that carbon ion beam irradiation in combination with immunotherapy yields better results, especially in controlling distant metastases. This implies that carbon ion induces a different anti-tumor immune response than photon beam. More complex molecular mechanisms need to be uncovered. This in vivo and in vitro experiment was carried out in order to examine the radio-immune effects and the mechanism of action of carbon ion beam versus X-ray in combination with PD-1 inhibitors. Methods and Materials: Lewis lung adenocarcinoma cells and C57BL/6 mice were used to create a tumor-bearing mouse model, with the non-irradiated tumor growing on the right hind leg and the irradiated tumor on the left rear. 10Gy carbon ion beam or X-ray radiation, either alone or in combination with PD-1 inhibitor, were used to treat the left back tumor. The expression of molecules linked to immunogenicity and the infiltration of CD8+ T lymphocytes into tumor tissues were both identified using immunohistochemistry. IFN-β in mouse serum was measured using an ELISA, while CD8+ T cells in mouse peripheral blood were measured using flow cytometry. Lewis cells were exposed to different dose of X-ray and carbon ion. TREX1, PD-L1, and IFN-β alterations in mRNA and protein levels were identified using Western blot or RT-PCR, respectively. TREX1 knockdown was created by siRNA transfection and exposed to various radiations. Using the CCK8 test, EdU assay, and flow cytometry, changes in cell viability, proliferation, and apoptosis rate were discovered. Results: Bilateral tumors were significantly inhibited by the use of carbon ion or X-ray in combination with PD-1, particularly to non-irradiated tumor(p<0.05). The percentage of infiltrating CD8+ T cells and the level of IFN-β expression were both raised by 10Gy carbon ion irradiation in the irradiated side tumor, although PD-L1 and TREX1 expression levels were also elevated. Lewis cell in vitro experiment further demonstrated that both X-ray and carbon ion irradiation can up-regulate the expression levels of PD-L1 and TREX1 with dose-dependent in tumors, particularly the trend of up-regulation TREX1 is more apparent at a higher dose in carbon ion, i.e. 8 or 10Gy, while the level of IFN-β is decreased. IFN-β levels were considerably raised under hypofractionated doses of carbon ion radiation by gene silencing TREX1. Conclusions: By enhancing tumor immunogenicity and increasing CD8+T infiltration in TME through a threshold dosage, X-ray or carbon ion radiation and PD-1 inhibitors improve anti-tumor activity and cause abscopal effect in Lewis lung adenocarcinoma-bearing mice. TREX1 is a possible therapeutic target and prognostic marker.
Collapse
Affiliation(s)
- Ruifeng Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Heavy Ion Medicine Center, Hospital of economic and Technological Development Zone, Wuhan Renmin Hospital of University, Wuhan, China
| | - Yichao Geng
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- The affiliated cancer hospital of Guizhou Medical University, Guiyang, China
| | - Hongtao Luo
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Qiuning Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Zhen Yang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Shan Li
- The affiliated cancer hospital of Guizhou Medical University, Guiyang, China
| | - Shilong Sun
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Zhiqiang Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Meng Dong
- The affiliated cancer hospital of Guizhou Medical University, Guiyang, China
| | - Tianqi Du
- The affiliated cancer hospital of Guizhou Medical University, Guiyang, China
| | - Tuanjie Che
- Key Laboratory of Functional Genomics and Molecular Diagnostics of Gansu Province, Lanzhou, China
| | - Xiaohu Wang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Key Laboratory of Functional Genomics and Molecular Diagnostics of Gansu Province, Lanzhou, China
| |
Collapse
|
11
|
Feng S, Luo H, Li C, Geng Y, Yang Z, Zhao X, Wang L, Liu R, Zhang Y, Che T, Zhang Q, Wang X. Regulatory role of RGMb in lung injury promoted by the combination of carbon ion irradiation and anti-PD-1 antibody through Erk1/2 and p38 MAPK pathways. Biochem Biophys Res Commun 2024; 691:149334. [PMID: 38042034 DOI: 10.1016/j.bbrc.2023.149334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 12/04/2023]
Abstract
The combination of carbon ion radiotherapy and anti-PD-1 antibody represents a new approach to treating thoracic tumors. However, the lung damage caused by this combination therapy may limit its use, and the potential mechanisms for this are worthy of investigation. The objective of this research was to examine the potential involvement of repulsive guidance molecule b (RGMb) in lung damage promoted by the utilization of carbon ion irradiation combined with an anti-PD-1 antibody. The C57BL/6 mice have been randomly separated into four distinct groups: control, anti-PD-1, whole thorax carbon ion irradiation, and irradiation in combination with anti-PD-1 treatment groups (combination group). Detection of pathological changes in lung tissue using HE staining. Detection of pulmonary fibrosis by Masson staining and the hydroxyproline assay. ELISA to detect TNF-α, TGF-β, IL-6, and IL-1β expression levels within lung homogenates. The expression of RGMb, p38 MAPK, and Erk1/2 pathways was detected using a fully automated digital Western blotting system WES (ProteinSimple, USA). Flow cytometry was employed to analyze tissue-resident memory T cells (TRM) within the lung. Subsequently, the siRNA gene was employed to induce the downregulation of RGMb in mice in order to validate the involvement of RGMb in radiation-immune lung injury. The present study observed a significant increase in both inflammatory and fibrotic indicators within the mice group's lung tissue that received the combination treatment. The combination group exhibited elevated levels of TGF-β, TNF-α, IL-6, and IL-1β in lung homogenates. Anti-PD-1 antibody and carbon ion irradiation, upregulated RGMb, phospho-p38 MAPK and phospho-Erk1/2. The results obtained from the flow cytometry analysis indicated that the combination group was significantly higher in the number of clonal expansion TRMs, which were predominantly characterized by the expression of CD8+CD103+CD69-TRMs. The downregulate of RGMb via siRNA in mice resulted in a decrease in phospho-p38 MAPK and phospho-Erk1/2. The combination group exhibited a reduction in TNF-α, TGF-β, IL-6, and IL-1β in their lung tissues, and the number of CD8+CD103+CD69-TRM was significantly reduced. The combination group exhibited a significant improvement in inflammatory and fibrotic indicators within the lung tissues. Anti-PD-1 antibody and carbon ion irradiation synergistically regulate RGMb, leading to strong clonal expansion of lung TRM through the p38 MAPK and Erk1/2 pathways. The present study offers valuable insights into the treatment of lung injury due to the combined administration of carbon ion radiotherapy and anti-PD-1 antibody therapy.
Collapse
Affiliation(s)
- Shuangwu Feng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
| | - Hongtao Luo
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Chengcheng Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
| | - Yichao Geng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
| | - Zhen Yang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China; Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China.
| | - Xueshan Zhao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
| | - Lina Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
| | - Ruifeng Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Yanying Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China.
| | - Tuanjie Che
- Key Laboratory of Functional Genomics and Molecular Diagnosis of Gansu Province, Lanzhou, Gansu, China.
| | - Qiuning Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Xiaohu Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China; Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China; University of Chinese Academy of Sciences, Beijing, China; Key Laboratory of Functional Genomics and Molecular Diagnosis of Gansu Province, Lanzhou, Gansu, China.
| |
Collapse
|
12
|
Helm A, Fournier C. High-LET charged particles: radiobiology and application for new approaches in radiotherapy. Strahlenther Onkol 2023; 199:1225-1241. [PMID: 37872399 PMCID: PMC10674019 DOI: 10.1007/s00066-023-02158-7] [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: 03/02/2023] [Accepted: 09/17/2023] [Indexed: 10/25/2023]
Abstract
The number of patients treated with charged-particle radiotherapy as well as the number of treatment centers is increasing worldwide, particularly regarding protons. However, high-linear energy transfer (LET) particles, mainly carbon ions, are of special interest for application in radiotherapy, as their special physical features result in high precision and hence lower toxicity, and at the same time in increased efficiency in cell inactivation in the target region, i.e., the tumor. The radiobiology of high-LET particles differs with respect to DNA damage repair, cytogenetic damage, and cell death type, and their increased LET can tackle cells' resistance to hypoxia. Recent developments and perspectives, e.g., the return of high-LET particle therapy to the US with a center planned at Mayo clinics, the application of carbon ion radiotherapy using cost-reducing cyclotrons and the application of helium is foreseen to increase the interest in this type of radiotherapy. However, further preclinical research is needed to better understand the differential radiobiological mechanisms as opposed to photon radiotherapy, which will help to guide future clinical studies for optimal exploitation of high-LET particle therapy, in particular related to new concepts and innovative approaches. Herein, we summarize the basics and recent progress in high-LET particle radiobiology with a focus on carbon ions and discuss the implications of current knowledge for charged-particle radiotherapy. We emphasize the potential of high-LET particles with respect to immunogenicity and especially their combination with immunotherapy.
Collapse
Affiliation(s)
- Alexander Helm
- Biophysics Department, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| | - Claudia Fournier
- Biophysics Department, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany.
| |
Collapse
|
13
|
Li P, Jia L, Bian X, Tan S. Application of Engineered Dendritic Cell Vaccines in Cancer Immunotherapy: Challenges and Opportunities. Curr Treat Options Oncol 2023; 24:1703-1719. [PMID: 37962824 DOI: 10.1007/s11864-023-01143-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2023] [Indexed: 11/15/2023]
Abstract
OPINION STATEMENT The primary objective of this study is to evaluate the effectiveness of cancer vaccines containing genetically modified dendritic cells (DCs) in inducing transformational immune responses. This paper sheds considerable light on DCs' function in advancing treatment techniques. This objective is achieved by thoroughly analyzing the many facets of DCs and their strategic integration into cancer treatment. Due to their role as immune response regulators, DCs can potentially enhance cancer treatment strategies. DCs have the potential to revolutionize immunotherapy, as shown by a comprehensive analysis of their numerous characteristics. The review deftly transitions from examining the fundamentals of preclinical research to delving into the complexities of clinical implementation while acknowledging the inherent challenges in translating DC vaccine concepts into tangible progress. The analysis also emphasizes the potential synergistic outcomes that can be achieved by combining DC vaccines with established pharmaceuticals, thereby emphasizing the importance of employing a holistic approach to enhance treatment efficacy. Despite the existence of transformative opportunities, advancement is hindered by several obstacles. The exhaustive analysis of technical complexities, regulatory dynamics, and upcoming challenges provides valuable insights for overcoming obstacles requiring strategic navigation to incorporate DC vaccines successfully. This document provides a comprehensive analysis of the developments in DC-based immunotherapy, concentrating on its potential to transform cancer therapy radically.
Collapse
Affiliation(s)
- Ping Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Linan Jia
- Department of Urology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Xiaobo Bian
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang110004, China
| | - Shutao Tan
- Department of Urology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, 110004, China.
| |
Collapse
|
14
|
Zhou Z, Guan B, Xia H, Zheng R, Xu B. Particle radiotherapy in the era of radioimmunotherapy. Cancer Lett 2023:216268. [PMID: 37331583 DOI: 10.1016/j.canlet.2023.216268] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/24/2023] [Accepted: 06/11/2023] [Indexed: 06/20/2023]
Abstract
Radiotherapy (RT) is one of the key modalities for cancer treatment, and more than 70% of tumor patients will receive RT during the course of their disease. Particle radiotherapy, such as proton radiotherapy, carbon-ion radiotherapy (CIRT) and boron neutron capture therapy (BNCT), is currently available for the treatment of patients Immunotherapy combined with photon RT has been successfully used in the clinic. The effect of immunotherapy combined with particle RT is an area of interest. However, the molecular mechanisms underlying the effects of combined immunotherapy and particle RT remain largely unknown. In this review, we summarize the properties of different types of particle RT and the mechanisms underlying their radiobiological effects. Additionally, we compared the main molecular players in photon RT and particle RT and the mechanisms involved the RT-mediated immune response.
Collapse
Affiliation(s)
- Zihan Zhou
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Bingjie Guan
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Huang Xia
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Rong Zheng
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China; Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China.
| | - Benhua Xu
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China; Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China.
| |
Collapse
|
15
|
Ainsworth V, Moreau M, Guthier R, Zegeye Y, Kozono D, Swanson W, Jandel M, Oh P, Quon H, Hobbs RF, Yasmin-Karim S, Sajo E, Ngwa W. Smart Radiotherapy Biomaterials for Image-Guided In Situ Cancer Vaccination. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1844. [PMID: 37368273 PMCID: PMC10303169 DOI: 10.3390/nano13121844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023]
Abstract
Recent studies have highlighted the potential of smart radiotherapy biomaterials (SRBs) for combining radiotherapy and immunotherapy. These SRBs include smart fiducial markers and smart nanoparticles made with high atomic number materials that can provide requisite image contrast during radiotherapy, increase tumor immunogenicity, and provide sustained local delivery of immunotherapy. Here, we review the state-of-the-art in this area of research, the challenges and opportunities, with a focus on in situ vaccination to expand the role of radiotherapy in the treatment of both local and metastatic disease. A roadmap for clinical translation is outlined with a focus on specific cancers where such an approach is readily translatable or will have the highest impact. The potential of FLASH radiotherapy to synergize with SRBs is discussed including prospects for using SRBs in place of currently used inert radiotherapy biomaterials such as fiducial markers, or spacers. While the bulk of this review focuses on the last decade, in some cases, relevant foundational work extends as far back as the last two and half decades.
Collapse
Affiliation(s)
- Victoria Ainsworth
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD 21201, USA; (M.M.); (H.Q.); (R.F.H.)
- Department of Physics, Medical Physics, University of Massachusetts Lowell, Lowell, MA 01854, USA (M.J.); (E.S.)
| | - Michele Moreau
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD 21201, USA; (M.M.); (H.Q.); (R.F.H.)
- Department of Physics, Medical Physics, University of Massachusetts Lowell, Lowell, MA 01854, USA (M.J.); (E.S.)
| | - Romy Guthier
- Department of Physics, Medical Physics, University of Massachusetts Lowell, Lowell, MA 01854, USA (M.J.); (E.S.)
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; (Y.Z.); (D.K.); (S.Y.-K.)
| | - Ysaac Zegeye
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; (Y.Z.); (D.K.); (S.Y.-K.)
- Department of Cell and Molecular Biology, Northeastern University, Boston, MA 02115, USA
| | - David Kozono
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; (Y.Z.); (D.K.); (S.Y.-K.)
| | - William Swanson
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA;
| | - Marian Jandel
- Department of Physics, Medical Physics, University of Massachusetts Lowell, Lowell, MA 01854, USA (M.J.); (E.S.)
| | - Philmo Oh
- NanoCan Therapeutics Corporation, Princeton, NJ 08540, USA;
| | - Harry Quon
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD 21201, USA; (M.M.); (H.Q.); (R.F.H.)
| | - Robert F. Hobbs
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD 21201, USA; (M.M.); (H.Q.); (R.F.H.)
| | - Sayeda Yasmin-Karim
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; (Y.Z.); (D.K.); (S.Y.-K.)
- Department of Radiation Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Erno Sajo
- Department of Physics, Medical Physics, University of Massachusetts Lowell, Lowell, MA 01854, USA (M.J.); (E.S.)
| | - Wilfred Ngwa
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD 21201, USA; (M.M.); (H.Q.); (R.F.H.)
- Department of Physics, Medical Physics, University of Massachusetts Lowell, Lowell, MA 01854, USA (M.J.); (E.S.)
| |
Collapse
|
16
|
Rutenberg MS, Beltran C. Future Perspective: Carbon Ion Radiotherapy for Head and Neck and Skull Base Malignancies. Oral Maxillofac Surg Clin North Am 2023:S1042-3699(23)00024-9. [PMID: 37117091 DOI: 10.1016/j.coms.2023.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Head and neck and base of skull malignancies are challenging for surgical and radiotherapy treatment due to the density of sensitive tissues. Carbon ion radiotherapy (CIRT) is a form of heavy particle therapy that uses accelerated carbon ions to treat malignancies that may be radioresistant or in challenging anatomic locations. CIRT has an increased biological effectiveness (ie, increased cell killing) at the end of the range of the carbon beam (ie, within the target tissue) but not in the entrance dose. This increased biological effectiveness can overcome the effects of radioresistant tumors, tissue hypoxia, and the need for radiotherapy fractionation.
Collapse
Affiliation(s)
- Michael S Rutenberg
- Department of Radiation Oncology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| | - Chris Beltran
- Department of Radiation Oncology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| |
Collapse
|
17
|
Sai S, Koto M, Yamada S. Basic and translational research on carbon-ion radiobiology. Am J Cancer Res 2023; 13:1-24. [PMID: 36777517 PMCID: PMC9906076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/16/2022] [Indexed: 02/14/2023] Open
Abstract
Carbon-ion beam irradiation (IR) has evident advantages over the conventional photon beams in treating tumors. It releases enormous amount of energy in a well-defined range with insignificant scatter in surrounding tissues based on well-localized energy deposition. Over the past 28 years, more than 14,000 patients with various types of cancer have been treated by carbon ion radiotherapy (CIRT) with promising results at QST. I have provided an overview of the basic and translational research on carbon-ion radiobiology including mechanisms underlying high linear energy transfer (LET) carbon-ion IR-induced cell death (apoptosis, autophagy, senescence, mitotic catastrophe etc.) and high radiocurability produced by carbon-ion beams in combination with DNA damaging drugs or with molecular-targeted drugs, micro-RNA therapeutics and immunotherapy. Additionally, I have focused on the application of these treatment in human cancer cells, especially cancer stem cells (CSCs). Finally, I have summarized the current studies on the application of basic carbon-ion beam IR according to the cancer types and clinical outcomes.
Collapse
Affiliation(s)
- Sei Sai
- Department of Charged Particle Therapy Research, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology (QST)Chiba, Japan
| | - Masashi Koto
- Department of Charged Particle Therapy Research, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology (QST)Chiba, Japan,QST Hospital, National Institutes for Quantum Science and Technology (QST)Chiba, Japan
| | - Shigeru Yamada
- QST Hospital, National Institutes for Quantum Science and Technology (QST)Chiba, Japan
| |
Collapse
|
18
|
Mirjolet C, Baude J, Galluzzi L. Dual impact of radiation therapy on tumor-targeting immune responses. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:xiii-xxiv. [PMID: 37438022 DOI: 10.1016/s1937-6448(23)00114-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Affiliation(s)
- Céline Mirjolet
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, GF Leclerc Centre, Unicancer, Dijon, France; TIReCS Team, UMR INSERM 1231, Dijon, France.
| | - Jérémy Baude
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, GF Leclerc Centre, Unicancer, Dijon, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| |
Collapse
|
19
|
Helm A, Totis C, Durante M, Fournier C. Are charged particles a good match for combination with immunotherapy? Current knowledge and perspectives. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 376:1-36. [PMID: 36997266 DOI: 10.1016/bs.ircmb.2023.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Charged particle radiotherapy, mainly using protons and carbon ions, provides physical characteristics allowing for a volume conformal irradiation and a reduction of the integral dose to normal tissue. Carbon ion therapy additionally features an increased biological effectiveness resulting in peculiar molecular effects. Immunotherapy, mostly performed with immune checkpoint inhibitors, is nowadays considered a pillar in cancer therapy. Based on the advantageous features of charged particle radiotherapy, we review pre-clinical evidence revealing a strong potential of its combination with immunotherapy. We argue that the combination therapy deserves further investigation with the aim of translation in clinics, where a few studies have been set up already.
Collapse
Affiliation(s)
- A Helm
- Biophysics Department, GSI, Darmstadt, Germany
| | - C Totis
- Biophysics Department, GSI, Darmstadt, Germany
| | - M Durante
- Biophysics Department, GSI, Darmstadt, Germany.
| | - C Fournier
- Biophysics Department, GSI, Darmstadt, Germany
| |
Collapse
|
20
|
Ovarian Cancer Radiosensitivity: What Have We Understood So Far? LIFE (BASEL, SWITZERLAND) 2022; 13:life13010006. [PMID: 36675955 PMCID: PMC9861683 DOI: 10.3390/life13010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/11/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
Radiotherapy has been increasingly considered as an active treatment to combine with other approaches (i.e., surgery, chemotherapy, and novel target-based drugs) in ovarian cancers to palliate symptoms and/or to prolong chemotherapy-free intervals. This narrative review aimed to summarize the current knowledge of the radiosensitivity/radioresistance of ovarian cancer which remains the most lethal gynecological cancer worldwide. Indeed, considering the high rate of recurrence in and out of the radiotherapy fields, in the era of patient-tailored oncology, elucidating the mechanisms of radiosensitivity and identifying potential radioresistance biomarkers could be crucial in guiding clinical decision-making.
Collapse
|
21
|
Biological Mechanisms to Reduce Radioresistance and Increase the Efficacy of Radiotherapy: State of the Art. Int J Mol Sci 2022; 23:ijms231810211. [PMID: 36142122 PMCID: PMC9499172 DOI: 10.3390/ijms231810211] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Cancer treatment with ionizing radiation (IR) is a well-established and effective clinical method to fight different types of tumors and is a palliative treatment to cure metastatic stages. Approximately half of all cancer patients undergo radiotherapy (RT) according to clinical protocols that employ two types of ionizing radiation: sparsely IR (i.e., X-rays) and densely IR (i.e., protons). Most cancer cells irradiated with therapeutic doses exhibit radio-induced cytotoxicity in terms of cell proliferation arrest and cell death by apoptosis. Nevertheless, despite the more tailored advances in RT protocols in the last few years, several tumors show a relatively high percentage of RT failure and tumor relapse due to their radioresistance. To counteract this extremely complex phenomenon and improve clinical protocols, several factors associated with radioresistance, of both a molecular and cellular nature, must be considered. Tumor genetics/epigenetics, tumor microenvironment, tumor metabolism, and the presence of non-malignant cells (i.e., fibroblast-associated cancer cells, macrophage-associated cancer cells, tumor-infiltrating lymphocytes, endothelial cells, cancer stem cells) are the main factors important in determining the tumor response to IR. Here, we attempt to provide an overview of how such factors can be taken advantage of in clinical strategies targeting radioresistant tumors.
Collapse
|
22
|
Tagliaferri L, Lancellotta V, Fionda B, Mangoni M, Casà C, Di Stefani A, Pagliara MM, D’Aviero A, Schinzari G, Chiesa S, Mazzarella C, Manfrida S, Colloca GF, Marazzi F, Morganti AG, Blasi MA, Peris K, Tortora G, Valentini V. Immunotherapy and radiotherapy in melanoma: a multidisciplinary comprehensive review. Hum Vaccin Immunother 2022; 18:1903827. [PMID: 33847208 PMCID: PMC9122308 DOI: 10.1080/21645515.2021.1903827] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/16/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Melanoma is an extremely aggressive tumor and is considered to be an extremely immunogenic tumor because compared to other cancers it usually presents a well-expressed lymphoid infiltration. The aim of this paper is to perform a multidisciplinary comprehensive review of the evidence available about the combination of radiotherapy and immunotherapy for melanoma. Radiation, in fact, can increase tumor antigens visibility and promote priming of T cells but can also exert immunosuppressive action on tumor microenvironment. Combining radiotherapy with immunotherapy provides an opportunity to increase immunostimulatory potential of radiation. We therefore provide the latest clinical evidence about radiobiological rationale, radiotherapy techniques, timing, and role both in advanced and systemic disease (with a special focus on ocular melanoma and brain, liver, and bone metastases) with a particular attention also in geriatric patients. The combination of immunotherapy and radiotherapy seems to be a safe therapeutic option, supported by a clear biological rationale, even though the available data confirm that radiotherapy is employed more for metastatic than for non-metastatic disease. Such a combination shows promising results in terms of survival outcomes; however, further studies, hopefully prospective, are needed to confirm such evidence.
Collapse
Affiliation(s)
- Luca Tagliaferri
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Valentina Lancellotta
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Bruno Fionda
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Monica Mangoni
- Sezione di Radioterapia Oncologica, Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università di Firenze, Florence, Italy
| | - Calogero Casà
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Alessandro Di Stefani
- UOC Dermatologia, Dipartimento di Scienze mediche e chirurgiche, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Monica Maria Pagliara
- UOC Oncologia Oculare, Dipartimento di Scienze dell'Invecchiamento, neurologiche ortopediche e della testa collo, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Andrea D’Aviero
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Giovanni Schinzari
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Silvia Chiesa
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Ciro Mazzarella
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Stefania Manfrida
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Giuseppe Ferdinando Colloca
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Fabio Marazzi
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Alessio Giuseppe Morganti
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale Settore Scientifico Disciplinare, Università di Bologna, Bologna, Italy
| | - Maria Antonietta Blasi
- UOC Oncologia Oculare, Dipartimento di Scienze dell'Invecchiamento, neurologiche ortopediche e della testa collo, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Ketty Peris
- UOC Dermatologia, Dipartimento di Scienze mediche e chirurgiche, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Giampaolo Tortora
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Vincenzo Valentini
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
23
|
Ma L. From Photon Beam to Accelerated Particle Beam: Antimetastasis Effect of Combining Radiotherapy With Immunotherapy. Front Public Health 2022; 10:847119. [PMID: 35425754 PMCID: PMC9002008 DOI: 10.3389/fpubh.2022.847119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/25/2022] [Indexed: 12/18/2022] Open
Abstract
Cancer is one of the major diseases that seriously threaten the human health. Radiotherapy is a common treatment for cancer. It is noninvasive and retains the functions of the organ where the tumor is located. Radiotherapy includes photon beam radiotherapy, which uses X-rays or gamma rays, and particle beam radiotherapy, using beams of protons and heavy ions. Compared with photon beam radiotherapy, particle beam radiotherapy has excellent dose distribution, which enables it to kill the primary tumor cells more effectively and simultaneously minimize the radiation-induced damage to normal tissues and organs surrounding the tumor. Despite the excellent therapeutic effect of particle beam radiotherapy on the irradiated tumors, it is not an effective treatment for metastatic cancers. Therefore, developing novel and effective treatment strategies for cancer is urgently needed to save patients with distant cancer metastasis. Immunotherapy enhances the body's own immune system to fight cancer by activating the immune cells, and consequently, to achieve the systemic anticancer effects, and it is considered to be an adjuvant therapy that can enhance the efficacy of particle beam radiotherapy. This review highlights the research progress of the antimetastasis effect and the mechanism of the photon beam or particle beam radiotherapy combined with immunotherapy and predicts the development prospects of this research area.
Collapse
Affiliation(s)
- Liqiu Ma
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing, China.,National Innovation Center of Radiation Application, Beijing, China
| |
Collapse
|
24
|
Particle radiotherapy and molecular therapies: mechanisms and strategies towards clinical applications. Expert Rev Mol Med 2022; 24:e8. [PMID: 35101155 DOI: 10.1017/erm.2022.2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Immunotherapy and targeted therapy are now commonly used in clinical trials in combination with radiotherapy for several cancers. While results are promising and encouraging, the molecular mechanisms of the interaction between the drugs and radiation remain largely unknown. This is especially important when switching from conventional photon therapy to particle therapy using protons or heavier ions. Different dose deposition patterns and molecular radiobiology can in fact modify the interaction with drugs and their effectiveness. We will show here that whilst the main molecular players are the same after low and high linear energy transfer radiation exposure, significant differences are observed in post-exposure signalling pathways that may lead to different effects of the drugs. We will also emphasise that the problem of the timing between drug administration and radiation and the fractionation regime are critical issues that need to be addressed urgently to achieve optimal results in combined treatments with particle therapy.
Collapse
|
25
|
Yu J, Sun H, Cao W, Song Y, Jiang Z. Research progress on dendritic cell vaccines in cancer immunotherapy. Exp Hematol Oncol 2022; 11:3. [PMID: 35074008 PMCID: PMC8784280 DOI: 10.1186/s40164-022-00257-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/16/2022] [Indexed: 12/13/2022] Open
Abstract
Dendritic cell (DC) vaccines induce specific immune responses that can selectively eliminate target cells. In recent years, many studies have been conducted to explore DC vaccination in the treatment of hematological malignancies, including acute myeloid leukemia and myelodysplastic syndromes, as well as other nonleukemia malignancies. There are at least two different strategies that use DCs to promote antitumor immunity: in situ vaccination and canonical vaccination. Monocyte-derived DCs (mo-DCs) and leukemia-derived DCs (DCleu) are the main types of DCs used in vaccines for AML and MDS thus far. Different cancer-related molecules such as peptides, recombinant proteins, apoptotic leukemic cells, whole tumor cells or lysates and DCs/DCleu containing a vaster antigenic repertoire with RNA electroporation, have been used as antigen sources to load DCs. To enhance DC vaccine efficacy, new strategies, such as combination with conventional chemotherapy, monospecific/bispecific antibodies and immune checkpoint-targeting therapies, have been explored. After a decade of trials and tribulations, much progress has been made and much promise has emerged in the field. In this review we summarize the recent advances in DC vaccine immunotherapy for AML/MDS as well as other nonleukemia malignancies.
Collapse
Affiliation(s)
- Jifeng Yu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan International Joint Laboratory of Nuclear Protein Gene Regulation, Henan University College of Medicine, Kaifeng, 475004, Henan, China
| | - Hao Sun
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Weijie Cao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yongping Song
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, Henan, China.
| | - Zhongxing Jiang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
26
|
Loap P, De Marzi L, Almeida CE, Barcellini A, Bradley J, de Santis MC, Dendale R, Jimenez R, Orlandi E, Kirova Y. Hadrontherapy techniques for breast cancer. Crit Rev Oncol Hematol 2021; 169:103574. [PMID: 34958916 DOI: 10.1016/j.critrevonc.2021.103574] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 12/31/2022] Open
Abstract
Radiotherapy plays a key role in breast cancer treatment, and recent technical advances have been made to improve the therapeutic window by limiting the risk of radiation-induced toxicity or by increasing tumor control. Hadrontherapy is a form a radiotherapy relying on particle beams; compared with photon beams, particle beams have specific physical, radiobiological and immunological properties, which can be valuable in diverse clinical situations. To date, available hadrontherapy techniques for breast cancer irradiation include proton therapy, carbon ion radiation therapy, fast neutron therapy and boron neutron capture therapy. This review analyzes the current rationale and level of evidence for each hadrontherapy technique for breast cancer.
Collapse
Affiliation(s)
- Pierre Loap
- Proton Therapy Center, Institut Curie, Orsay, France.
| | | | - Carlos Eduardo Almeida
- Department of Radiological Sciences, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Julie Bradley
- University of Florida Health Proton Therapy Institute, Jacksonville, FL, United States
| | | | - Remi Dendale
- Proton Therapy Center, Institut Curie, Orsay, France
| | - Rachel Jimenez
- Massachusetts General Hospital, Boston, MA, United States
| | - Ester Orlandi
- National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Youlia Kirova
- Proton Therapy Center, Institut Curie, Orsay, France
| |
Collapse
|
27
|
Loap P, Vischioni B, Bonora M, Ingargiola R, Ronchi S, Vitolo V, Barcellini A, Goanta L, De Marzi L, Dendale R, Pacelli R, Locati L, Calugaru V, Mammar H, Cavalieri S, Kirova Y, Orlandi E. Biological Rationale and Clinical Evidence of Carbon Ion Radiation Therapy for Adenoid Cystic Carcinoma: A Narrative Review. Front Oncol 2021; 11:789079. [PMID: 34917512 PMCID: PMC8668942 DOI: 10.3389/fonc.2021.789079] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Adenoid cystic carcinoma (ACC) is a rare, basaloid, epithelial tumor, arising mostly from salivary glands. Radiation therapy can be employed as a single modality for unresectable tumors, in an adjuvant setting after uncomplete resection, in case of high-risk pathological features, or for recurrent tumors. Due to ACC intrinsic radioresistance, high linear energy transfer (LET) radiotherapy techniques have been evaluated for ACC irradiation: while fast neutron therapy has now been abandoned due to toxicity concerns, charged particle beams such as protons and carbon ions are at present the beams used for hadron therapy. Carbon ion radiation therapy (CIRT) is currently increasingly used for ACC irradiation. The aim of this review is to describe the immunological, molecular and clinicopathological bases that support ACC treatment with CIRT, as well as to expose the current clinical evidence that reveal the advantages of using CIRT for treating ACC.
Collapse
Affiliation(s)
- Pierre Loap
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy.,Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France
| | - Barbara Vischioni
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Maria Bonora
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Rossana Ingargiola
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Sara Ronchi
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Viviana Vitolo
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Amelia Barcellini
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Lucia Goanta
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Napoli, Italy
| | - Ludovic De Marzi
- Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France.,Institut Curie, PSL Research University, University Paris Saclay, INSERM LITO, Orsay, France
| | - Remi Dendale
- Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France
| | - Roberto Pacelli
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Napoli, Italy
| | - Laura Locati
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Valentin Calugaru
- Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France
| | - Hamid Mammar
- Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France
| | - Stefano Cavalieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Youlia Kirova
- Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France
| | - Ester Orlandi
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| |
Collapse
|
28
|
Zhang P, Meng J, Li Y, Yang C, Hou Y, Tang W, McHugh KJ, Jing L. Nanotechnology-enhanced immunotherapy for metastatic cancer. Innovation (N Y) 2021; 2:100174. [PMID: 34766099 PMCID: PMC8571799 DOI: 10.1016/j.xinn.2021.100174] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
A vast majority of cancer deaths occur as a result of metastasis. Unfortunately, effective treatments for metastases are currently lacking due to the difficulty of selectively targeting these small, delocalized tumors distributed across a variety of organs. However, nanotechnology holds tremendous promise for improving immunotherapeutic outcomes in patients with metastatic cancer. In contrast to conventional cancer immunotherapies, rationally designed nanomaterials can trigger specific tumoricidal effects, thereby improving immune cell access to major sites of metastasis such as bone, lungs, and lymph nodes, optimizing antigen presentation, and inducing a persistent immune response. This paper reviews the cutting-edge trends in nano-immunoengineering for metastatic cancers with an emphasis on different nano-immunotherapeutic strategies. Specifically, it discusses directly reversing the immunological status of the primary tumor, harnessing the potential of peripheral immune cells, preventing the formation of a pre-metastatic niche, and inhibiting the tumor recurrence through postoperative immunotherapy. Finally, we describe the challenges facing the integration of nanoscale immunomodulators and provide a forward-looking perspective on the innovative nanotechnology-based tools that may ultimately prove effective at eradicating metastatic diseases.
Collapse
Affiliation(s)
- Peisen Zhang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Junli Meng
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Yingying Li
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Chen Yang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Yi Hou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Wen Tang
- South China Advanced Institute for Soft Matter Science and Technology, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, 6100 Main Street, MS-142, Houston, TX 77005, USA
| | - Lihong Jing
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| |
Collapse
|
29
|
Ma L, Sakamoto Y, Ando K, Fujita H, Takahashi A, Takeshima T, Otsuka H, Ebner DK, Kakimi K, Imai T, Shimokawa T. Th balance related host genetic background affects the therapeutic effects of combining carbon-ion radiotherapy with dendritic cell immunotherapy. Int J Radiat Oncol Biol Phys 2021; 112:780-789. [PMID: 34740767 DOI: 10.1016/j.ijrobp.2021.10.141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE The goal of this study is to clarify the underlying mechanisms of metastasis suppression by CiDC (carbon-ion radiotherapy (CIRT) combined with immature dendritic cell (iDC) immunotherapy), which was previously shown to significantly suppress pulmonary metastasis in a NR-S1-bearing C3H/He mouse model. METHODS AND MATERIALS Mouse carcinoma cell lines (LLC, LM8, Colon-26 and Colon-26MGS) were grafted into the right hind paw of syngeneic mice (C57BL/6J, C3H/He and BALB/c). Seven days later, the tumors on the mice were locally irradiated with carbon-ions (290 MeV/n, 6 cm SOBP, 1 or 2 Gy). At 1.5 days after irradiation, bone marrow-derived immature dendritic cells were administrated intravenously into a subset of the mice. The number of lung metastases was evaluated within three weeks after irradiation. In vitro cultured cancer cells were irradiated with carbon-ion (290 MeV/n, mono-energy, LET approximately 70 ∼ 80 keV/µm), and then co-cultured with iDCs for three days to determine the DC maturation. RESULTS CiDC effectively repressed distant lung metastases in cancer cell (LLC and LM8)-bearing C57BL/6J and C3H/He mouse models. However, Colon-26 and Colon-26MGS-bearing BALB/c models did not show enhancement of metastasis suppression by combination treatment. This was further evaluated by comparing LM8-bearing C3H/He and LLC-bearing C57BL/6J models with a Colon-26-bearing BALB/c model. In vitro co-culture assays demonstrated that all irradiated cell lines were able to activate C3H/He or C57BL/6J-derived iDCs into mature DCs, but not BALB/c-derived iDCs. CONCLUSION The genetic background of the host may have a strong impact on the potency of combination therapy. Future animal and clinical testing should evaluate host genetic factors when evaluating treatment efficacy.
Collapse
Affiliation(s)
- Liqiu Ma
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan; Gunma University Heavy Ion Medical Center, Gunma, Japan; China Institute of Atomic Energy, Beijing 102413, China
| | - Yoshimitsu Sakamoto
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ken Ando
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan; Department of Radiation Oncology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hidetoshi Fujita
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | | | - Tsuguhide Takeshima
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hiromi Otsuka
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Daniel K Ebner
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo, Tokyo, Japan
| | - Takashi Imai
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takashi Shimokawa
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.
| |
Collapse
|
30
|
Averbeck D, Rodriguez-Lafrasse C. Role of Mitochondria in Radiation Responses: Epigenetic, Metabolic, and Signaling Impacts. Int J Mol Sci 2021; 22:ijms222011047. [PMID: 34681703 PMCID: PMC8541263 DOI: 10.3390/ijms222011047] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Until recently, radiation effects have been considered to be mainly due to nuclear DNA damage and their management by repair mechanisms. However, molecular biology studies reveal that the outcomes of exposures to ionizing radiation (IR) highly depend on activation and regulation through other molecular components of organelles that determine cell survival and proliferation capacities. As typical epigenetic-regulated organelles and central power stations of cells, mitochondria play an important pivotal role in those responses. They direct cellular metabolism, energy supply and homeostasis as well as radiation-induced signaling, cell death, and immunological responses. This review is focused on how energy, dose and quality of IR affect mitochondria-dependent epigenetic and functional control at the cellular and tissue level. Low-dose radiation effects on mitochondria appear to be associated with epigenetic and non-targeted effects involved in genomic instability and adaptive responses, whereas high-dose radiation effects (>1 Gy) concern therapeutic effects of radiation and long-term outcomes involving mitochondria-mediated innate and adaptive immune responses. Both effects depend on radiation quality. For example, the increased efficacy of high linear energy transfer particle radiotherapy, e.g., C-ion radiotherapy, relies on the reduction of anastasis, enhanced mitochondria-mediated apoptosis and immunogenic (antitumor) responses.
Collapse
Affiliation(s)
- Dietrich Averbeck
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France;
- Correspondence:
| | - Claire Rodriguez-Lafrasse
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France;
- Department of Biochemistry and Molecular Biology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| |
Collapse
|
31
|
Wang J, Dai Z, Miao Y, Zhao T, Gan J, Zhao C, Ran J, Guan Q. Carbon ion ( 12C 6+) irradiation induces the expression of Klrk1 in lung cancer and optimizes the tumor microenvironment based on the NKG2D/NKG2D-Ls pathway. Cancer Lett 2021; 521:178-195. [PMID: 34492331 DOI: 10.1016/j.canlet.2021.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022]
Abstract
With the identification of "negative immune regulation" defects in the immune system and the continuous improvement of immunotherapy, natural killer cells (NK) have received more attention, especially as tools in combined immunotherapy. Carbon ions (12C6+) have become the ideal radiation for combined immunotherapy due to their significant radiobiological advantages and synergistic effects. The purpose of this study was to explore the NK cell-mediated cytotoxicity pathway and related mechanisms in lung cancer induced by carbon ion irradiation. KLRK1, which specifically encodes the NKG2D receptor, was significantly correlated with the prognosis, clinical stage, functional status of NK cells, and the immune microenvironment of lung cancer, as shown by bioinformatics analysis. Based on RNA-seq data of Lewis lung cancer in C57BL/6 mice, carbon ion irradiation was found to significantly induce Klrk1 gene expression and activate the NKG2D/NKG2D-Ls pathway. The Treg inhibition pathway combined with carbon ion radiotherapy could significantly increase the infiltration and function of NK cells in the tumor microenvironment of lung cancer and prolong the survival time of C57BL/6 mice. In conclusion, carbon ions have significant radiobiological advantages, especially under conditions of combined immunotherapy. Carbon ions combined with Treg inhibitors can significantly improve the infiltration and functional status of NK cells.
Collapse
Affiliation(s)
- Jiangtao Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, PR China
| | - Ziying Dai
- Department of Radiation Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, PR China
| | - Yandong Miao
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, PR China
| | - Ting Zhao
- Medical Physics Room, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, PR China
| | - Jian Gan
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, PR China
| | - Chengpeng Zhao
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, PR China
| | - Juntao Ran
- Department of Radiation Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, PR China.
| | - Quanlin Guan
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, PR China; Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, PR China.
| |
Collapse
|
32
|
Matsumoto Y, Fukumitsu N, Ishikawa H, Nakai K, Sakurai H. A Critical Review of Radiation Therapy: From Particle Beam Therapy (Proton, Carbon, and BNCT) to Beyond. J Pers Med 2021; 11:jpm11080825. [PMID: 34442469 PMCID: PMC8399040 DOI: 10.3390/jpm11080825] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/20/2021] [Accepted: 08/22/2021] [Indexed: 12/24/2022] Open
Abstract
In this paper, we discuss the role of particle therapy—a novel radiation therapy (RT) that has shown rapid progress and widespread use in recent years—in multidisciplinary treatment. Three types of particle therapies are currently used for cancer treatment: proton beam therapy (PBT), carbon-ion beam therapy (CIBT), and boron neutron capture therapy (BNCT). PBT and CIBT have been reported to have excellent therapeutic results owing to the physical characteristics of their Bragg peaks. Variable drug therapies, such as chemotherapy, hormone therapy, and immunotherapy, are combined in various treatment strategies, and treatment effects have been improved. BNCT has a high dose concentration for cancer in terms of nuclear reactions with boron. BNCT is a next-generation RT that can achieve cancer cell-selective therapeutic effects, and its effectiveness strongly depends on the selective 10B accumulation in cancer cells by concomitant boron preparation. Therefore, drug delivery research, including nanoparticles, is highly desirable. In this review, we introduce both clinical and basic aspects of particle beam therapy from the perspective of multidisciplinary treatment, which is expected to expand further in the future.
Collapse
Affiliation(s)
- Yoshitaka Matsumoto
- Department of Radiation Oncology, Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; (K.N.); (H.S.)
- Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba 305-8576, Japan
- Correspondence: ; Tel.: +81-29-853-7100
| | | | - Hitoshi Ishikawa
- National Institute of Quantum and Radiological Science and Technology Hospital, Chiba 263-8555, Japan;
| | - Kei Nakai
- Department of Radiation Oncology, Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; (K.N.); (H.S.)
- Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba 305-8576, Japan
| | - Hideyuki Sakurai
- Department of Radiation Oncology, Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; (K.N.); (H.S.)
- Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba 305-8576, Japan
| |
Collapse
|
33
|
Byun HK, Han MC, Yang K, Kim JS, Yoo GS, Koom WS, Kim YB. Physical and Biological Characteristics of Particle Therapy for Oncologists. Cancer Res Treat 2021; 53:611-620. [PMID: 34139805 PMCID: PMC8291193 DOI: 10.4143/crt.2021.066] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
Particle therapy is a promising and evolving modality of radiotherapy that can be used to treat tumors that are radioresistant to conventional photon beam radiotherapy. It has unique biological and physical advantages compared with conventional radiotherapy. The characteristic feature of particle therapy is the "Bragg peak," a steep and localized peak of dose, that enables precise delivery of the radiation dose to the tumor while effectively sparing normal organs. Especially, the charged particles (e.g., proton, helium, carbon) cause a high rate of energy loss along the track, thereby leading to high biological effectiveness, which makes particle therapy attractive. Using this property, the particle beam induces more severe DNA double-strand breaks than the photon beam, which is less influenced by the oxygen level. This review describes the general biological and physical aspects of particle therapy for oncologists, including non-radiation oncologists and beginners in the field.
Collapse
Affiliation(s)
- Hwa Kyung Byun
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Min Cheol Han
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Kyungmi Yang
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Sung Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Gyu Sang Yoo
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Bae Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
34
|
Marcus D, Lieverse RIY, Klein C, Abdollahi A, Lambin P, Dubois LJ, Yaromina A. Charged Particle and Conventional Radiotherapy: Current Implications as Partner for Immunotherapy. Cancers (Basel) 2021; 13:1468. [PMID: 33806808 PMCID: PMC8005048 DOI: 10.3390/cancers13061468] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy (RT) has been shown to interfere with inflammatory signals and to enhance tumor immunogenicity via, e.g., immunogenic cell death, thereby potentially augmenting the therapeutic efficacy of immunotherapy. Conventional RT consists predominantly of high energy photon beams. Hypofractionated RT regimens administered, e.g., by stereotactic body radiation therapy (SBRT), are increasingly investigated in combination with cancer immunotherapy within clinical trials. Despite intensive preclinical studies, the optimal dose per fraction and dose schemes for elaboration of RT induced immunogenic potential remain inconclusive. Compared to the scenario of combined immune checkpoint inhibition (ICI) and RT, multimodal therapies utilizing other immunotherapy principles such as adoptive transfer of immune cells, vaccination strategies, targeted immune-cytokines and agonists are underrepresented in both preclinical and clinical settings. Despite the clinical success of ICI and RT combination, e.g., prolonging overall survival in locally advanced lung cancer, curative outcomes are still not achieved for most cancer entities studied. Charged particle RT (PRT) has gained interest as it may enhance tumor immunogenicity compared to conventional RT due to its unique biological and physical properties. However, whether PRT in combination with immune therapy will elicit superior antitumor effects both locally and systemically needs to be further investigated. In this review, the immunological effects of RT in the tumor microenvironment are summarized to understand their implications for immunotherapy combinations. Attention will be given to the various immunotherapeutic interventions that have been co-administered with RT so far. Furthermore, the theoretical basis and first evidences supporting a favorable immunogenicity profile of PRT will be examined.
Collapse
Affiliation(s)
- Damiënne Marcus
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Relinde I. Y. Lieverse
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Carmen Klein
- German Cancer Consortium (DKTK) Core-Center Heidelberg, National Center for Tumor Diseases (NCT), Clinical Cooperation Unit Translational Radiation Oncology, Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120 Heidelberg, Germany; (C.K.); (A.A.)
- Heidelberg Ion-Beam Therapy Center (HIT), Division of Molecular and Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
- National Center for Radiation Oncology (NCRO), Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 222, 69120 Heidelberg, Germany
| | - Amir Abdollahi
- German Cancer Consortium (DKTK) Core-Center Heidelberg, National Center for Tumor Diseases (NCT), Clinical Cooperation Unit Translational Radiation Oncology, Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120 Heidelberg, Germany; (C.K.); (A.A.)
- Heidelberg Ion-Beam Therapy Center (HIT), Division of Molecular and Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
- National Center for Radiation Oncology (NCRO), Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 222, 69120 Heidelberg, Germany
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Ludwig J. Dubois
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| |
Collapse
|
35
|
Guo T, Zou L, Ni J, Chu X, Zhu Z. Radiotherapy for unresectable locally advanced non-small cell lung cancer: a narrative review of the current landscape and future prospects in the era of immunotherapy. Transl Lung Cancer Res 2020; 9:2097-2112. [PMID: 33209629 PMCID: PMC7653144 DOI: 10.21037/tlcr-20-511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significant recent advances have occurred in the use of radiation therapy for locally advanced non-small cell lung cancer (LA-NSCLC). In fact, the past few decades have seen both therapeutic gains and setbacks in the evolution of radiotherapy for LA-NSCLC. The PACIFIC trial has heralded a new era of immunotherapy and has raised important questions for future study, such as the future directions of radiation therapy for LA-NSCLC in the era of immunotherapy. Modern radiotherapy techniques such as three-dimensional (3D) conformal radiotherapy and intensity-modulated radiotherapy (IMRT) provide opportunities for improved target conformity and reduced normal-tissue exposure. However, the low-dose radiation volume brought by IMRT and its effects on the immune system deserve particular attention when combing radiotherapy and immunotherapy. Particle radiotherapy offers dosimetric advantages and exhibits great immunoregulatory potential. With the ongoing improvement in particle radiotherapy techniques and knowledge, the combination of immunotherapy and particle radiotherapy has tremendous potential to improve treatment outcomes. Of particular importance are questions on the optimal radiation schedule in the settings of radio-immunotherapy. Strategies for the reduction of the irradiated field such as involved-field irradiation (IFI) and omission of clinical target volume (CTV) hold promise for better preservation of immune function while not compromising locoregional and distant control. In addition, different dose-fractionation regimens can have diverse effects on the immune system. Thus, prospective trials are urgently needed to establish the optimal dose fractionation regimen. Moreover, personalized radiotherapy which allows the tailoring of radiation dose to each individual's genetic background and immune state is of critical importance in maximizing the benefit of radiation to patients with LA-NSCLC.
Collapse
Affiliation(s)
- Tiantian Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College
| | - Liqing Zou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College
| | - Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College
| | - Xiao Chu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College.,Institute of Thoracic Oncology, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Tinganelli W, Durante M. Carbon Ion Radiobiology. Cancers (Basel) 2020; 12:E3022. [PMID: 33080914 PMCID: PMC7603235 DOI: 10.3390/cancers12103022] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
Radiotherapy using accelerated charged particles is rapidly growing worldwide. About 85% of the cancer patients receiving particle therapy are irradiated with protons, which have physical advantages compared to X-rays but a similar biological response. In addition to the ballistic advantages, heavy ions present specific radiobiological features that can make them attractive for treating radioresistant, hypoxic tumors. An ideal heavy ion should have lower toxicity in the entrance channel (normal tissue) and be exquisitely effective in the target region (tumor). Carbon ions have been chosen because they represent the best combination in this direction. Normal tissue toxicities and second cancer risk are similar to those observed in conventional radiotherapy. In the target region, they have increased relative biological effectiveness and a reduced oxygen enhancement ratio compared to X-rays. Some radiobiological properties of densely ionizing carbon ions are so distinct from X-rays and protons that they can be considered as a different "drug" in oncology, and may elicit favorable responses such as an increased immune response and reduced angiogenesis and metastatic potential. The radiobiological properties of carbon ions should guide patient selection and treatment protocols to achieve optimal clinical results.
Collapse
Affiliation(s)
- Walter Tinganelli
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforchung, Planckstraße 1, 64291 Darmstadt, Germany;
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforchung, Planckstraße 1, 64291 Darmstadt, Germany;
- Institut für Festkörperphysik, Technische Universität Darmstadt, Hochschulstraße 8, 64289 Darmstadt, Germany
| |
Collapse
|
37
|
Helm A, Tinganelli W, Simoniello P, Kurosawa F, Fournier C, Shimokawa T, Durante M. Reduction of Lung Metastases in a Mouse Osteosarcoma Model Treated With Carbon Ions and Immune Checkpoint Inhibitors. Int J Radiat Oncol Biol Phys 2020; 109:594-602. [PMID: 32980497 DOI: 10.1016/j.ijrobp.2020.09.041] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 08/12/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE The combination of radiation therapy and immunotherapy is recognized as a very promising strategy for metastatic cancer treatment. The purpose of this work is to compare the effectiveness of x-ray and high-energy carbon ion therapy in combination with checkpoint inhibitors in a murine model. METHODS AND MATERIALS We used an osteosarcoma mouse model irradiated with either carbon ions or x-rays in combination with 2 immune checkpoint inhibitors (anti-PD-1 and anti-CTLA-4). LM8 osteosarcoma cells were injected in both hind limbs of female C3H/He mice 7 days before exposure to carbon ions or x-rays. In experimental groups receiving irradiation, only the tumor on the left limb was exposed, whereas the tumor on the right limb served as an abscopal mimic. Checkpoint inhibitors were injected intraperitoneally 1 day before exposure as well as concomitant to and after exposure. Tumor growth was measured regularly up to day 21 after exposure, when mice were sacrificed. Both tumors as well as lungs were extracted. RESULTS A reduced growth of the abscopal tumor was most pronounced after the combined protocol of carbon ions and the immune checkpoint inhibitors administered sequentially. Radiation or checkpoint inhibitors alone were not sufficient to reduce the growth of the abscopal tumors. Carbon ions alone reduced the number of lung metastases more efficiently than x-rays, and in combination with immunotherapy both radiation types essentially suppressed the metastasis, with carbon ions being again more efficient. Investigation of the infiltration of immune cells in the abscopal tumors of animals treated with combination revealed an increase in CD8+ cells. CONCLUSIONS Combination of checkpoint inhibitors with high-energy carbon ion radiation therapy can be an effective strategy for the treatment of advanced tumors.
Collapse
Affiliation(s)
- Alexander Helm
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Walter Tinganelli
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Palma Simoniello
- Department of Science and Technology, Parthenope University of Naples, Naples, Italy; Istituto Nazionale di Fisica Nucleare, Sezione di Napoli, Naples, Italy
| | - Fuki Kurosawa
- National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| | - Claudia Fournier
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Takashi Shimokawa
- National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| | - Marco Durante
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany; Technische Universität Darmstadt, Institut für Festkörperphysik, Darmstadt, Germany.
| |
Collapse
|
38
|
Gómez V, Mustapha R, Ng K, Ng T. Radiation therapy and the innate immune response: Clinical implications for immunotherapy approaches. Br J Clin Pharmacol 2020; 86:1726-1735. [PMID: 32388875 PMCID: PMC7444780 DOI: 10.1111/bcp.14351] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy is an essential component of cancer care, contributing up to 40% of curative cancer treatment regimens. It creates DNA double-strand breaks causing cell death in highly replicating tumour cells. However, tumours can develop acquired resistance to therapy. The efficiency of radiation treatment has been increased by means of combining it with other approaches such as chemotherapy, molecule-targeted therapies and, in recent years, immunotherapy (IT). Cancer-cell apoptosis after radiation treatment causes an immunological reaction that contributes to eradicating the tumour via antigen presentation and subsequent T-cell activation. By contrast, radiotherapy also contributes to the formation of an immunosuppressive environment that hinders the efficacy of the therapy. Innate immune cells from myeloid and lymphoid origin show a very active role in both acquired resistance and antitumourigenic mechanisms. Therefore, many efforts are being made in order to reach a better understanding of the innate immunity reactions after radiation therapy (RT) and the design of new combinatorial IT strategies focused in these particular populations.
Collapse
Affiliation(s)
- Valentí Gómez
- UCL Cancer InstituteUniversity College LondonLondonUK
- Cancer Research UK City of London CentreUK
| | - Rami Mustapha
- School of Cancer and Pharmaceutical SciencesKing's College LondonLondonUK
- Cancer Research UK King's Health Partners CentreUK
| | - Kenrick Ng
- UCL Cancer InstituteUniversity College LondonLondonUK
- Department of Medical OncologyUniversity College Hospitals NHS Foundation TrustUK
| | - Tony Ng
- UCL Cancer InstituteUniversity College LondonLondonUK
- Cancer Research UK City of London CentreUK
- School of Cancer and Pharmaceutical SciencesKing's College LondonLondonUK
- Cancer Research UK King's Health Partners CentreUK
| |
Collapse
|
39
|
Malouff TD, Mahajan A, Krishnan S, Beltran C, Seneviratne DS, Trifiletti DM. Carbon Ion Therapy: A Modern Review of an Emerging Technology. Front Oncol 2020; 10:82. [PMID: 32117737 PMCID: PMC7010911 DOI: 10.3389/fonc.2020.00082] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/16/2020] [Indexed: 12/13/2022] Open
Abstract
Radiation therapy is one of the most widely used therapies for malignancies. The therapeutic use of heavy ions, such as carbon, has gained significant interest due to advantageous physical and radiobiologic properties compared to photon based therapy. By taking advantage of these unique properties, carbon ion radiotherapy may allow dose escalation to tumors while reducing radiation dose to adjacent normal tissues. There are currently 13 centers treating with carbon ion radiotherapy, with many of these centers publishing promising safety and efficacy data from the first cohorts of patients treated. To date, carbon ion radiotherapy has been studied for almost every type of malignancy, including intracranial malignancies, head and neck malignancies, primary and metastatic lung cancers, tumors of the gastrointestinal tract, prostate and genitourinary cancers, sarcomas, cutaneous malignancies, breast cancer, gynecologic malignancies, and pediatric cancers. Additionally, carbon ion radiotherapy has been studied extensively in the setting of recurrent disease. We aim to provide a comprehensive review of the studies of each of these disease sites, with a focus on the current trials using carbon ion radiotherapy.
Collapse
|
40
|
Vitolo V, Cobianchi L, Brugnatelli S, Barcellini A, Peloso A, Facoetti A, Vanoli A, Delfanti S, Preda L, Molinelli S, Klersy C, Fossati P, Orecchia R, Valvo F. Preoperative chemotherapy and carbon ions therapy for treatment of resectable and borderline resectable pancreatic adenocarcinoma: a prospective, phase II, multicentre, single-arm study. BMC Cancer 2019; 19:922. [PMID: 31521134 PMCID: PMC6744648 DOI: 10.1186/s12885-019-6108-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/29/2019] [Indexed: 12/17/2022] Open
Abstract
Background Pancreatic adenocarcinoma is a high-mortality neoplasm with a documented 5-years-overall survival around 5%. In the last decades, a real breakthrough in the treatment of the disease has not been achieved. Here we propose a prospective, phase II, multicentre, single-arm study aiming to assess the efficacy and the feasibility of a therapeutic protocol combining chemotherapy, carbon ion therapy and surgery for resectable and borderline resectable pancreatic adenocarcinoma. Method The purpose of this trial (PIOPPO Protocol) is to assess the efficacy and the feasibility of 3 cycles of FOLFIRINOX neoadjuvant chemotherapy followed by a short-course of carbon ion radiotherapy (CIRT) for resectable or borderline resectable pancreatic adenocarcinoma patients. Primary outcome of this study is the assessment of local progression free survival (L-PFS). The calculation of sample size is based on the analysis of the primary endpoint “progression free survival” according to Fleming’s Procedure. Discussion Very preliminary results provide initial evidence of the feasibility of the combined chemotherapy and CIRT in the neoadjuvant setting for resectable or borderline resectable pancreatic cancer. Completion of the accrual and long term results are awaited to see if this combination of treatment is advisable and will provide the expected benefits. Trial registration ClinicalTrials.gov Identifier: NCT03822936 registered on January 2019.
Collapse
Affiliation(s)
- Viviana Vitolo
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| | - Lorenzo Cobianchi
- General Surgery Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Silvia Brugnatelli
- Department of Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Amelia Barcellini
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy.
| | - Andrea Peloso
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Divisions of Abdominal and Transplantation Surgery, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Angelica Facoetti
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Sara Delfanti
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Lorenzo Preda
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy.,Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Silvia Molinelli
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| | - Catherine Klersy
- Service of Clinical Epidemiology & Biometry, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Piero Fossati
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy.,MedAustron Ion Therapy Center, Wiener Neustadt, Austria
| | - Roberto Orecchia
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy.,European Institute of Oncology (IEO), Milan, Italy
| | - Francesca Valvo
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| |
Collapse
|
41
|
Durante M, Formenti S. Harnessing radiation to improve immunotherapy: better with particles? Br J Radiol 2019; 93:20190224. [PMID: 31317768 DOI: 10.1259/bjr.20190224] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The combination of radiotherapy and immunotherapy is one of the most promising strategies for cancer treatment. Recent clinical results support the pre-clinical experiments pointing to a benefit for the combined treatment in metastatic patients. Charged particle therapy (using protons or heavier ions) is considered one of the most advanced radiotherapy techniques, but its cost remains higher than conventional X-ray therapy. The most important question to be addressed to justify a more widespread use of particle therapy is whether they can be more effective than X-rays in combination with immunotherapy. Protons and heavy ions have physical advantages compared to X-rays that lead to a reduced damage to the immune cells, that are required for an effective immune response. Moreover, densely ionizing radiation may have biological advantages, due to different cell death pathways and release of cytokine mediators of inflammation. We will discuss results in esophageal cancer patients showing that charged particles can reduce the damage to blood lymphocytes compared to X-rays, and preliminary in vitro studies pointing to an increased release of immune-stimulating cytokines after heavy ion exposure. Pre-clinical and clinical studies are ongoing to test these hypotheses.
Collapse
Affiliation(s)
- Marco Durante
- GSI Helmholtzzentrum für Schwerionenforschung, Biophysics Department, Darmstadt, Germany.,Technische Universität Darmstadt, Institut für Festkörperphysik, Darmstadt, Germany
| | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
42
|
Mohsenzadegan M, Peng RW, Roudi R. Dendritic cell/cytokine-induced killer cell-based immunotherapy in lung cancer: What we know and future landscape. J Cell Physiol 2019; 235:74-86. [PMID: 31222740 DOI: 10.1002/jcp.28977] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/28/2019] [Accepted: 05/31/2019] [Indexed: 12/11/2022]
Abstract
Multiple modalities for lung cancer therapy have emerged in the past decade, whereas their clinical applications and survival-beneficiary is little known. Vaccination with dendritic cells (DCs) or DCs/cytokine-induced killer (CIK) cells has shown limited success in the treatment of patients with advanced non-small-cell lung cancer. To evaluate and overcome these limitations in further studies, in the present review, we sum up recent progress about DCs or DCs/CIKs-based approaches for preclinical and clinical trials in patients with lung cancer and discuss some of the limited therapeutic success. Moreover, this review highlights the need to focus future studies on the development of new approaches for successful immunotherapy in patients with lung cancer.
Collapse
Affiliation(s)
- Monireh Mohsenzadegan
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Ren-Wang Peng
- Division of General Thoracic Surgery, Department for BioMedical Research (DBMR), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Raheleh Roudi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Kong L, Gao J, Hu J, Lu R, Yang J, Qiu X, Hu W, Lu JJ. Carbon ion radiotherapy boost in the treatment of glioblastoma: a randomized phase I/III clinical trial. Cancer Commun (Lond) 2019; 39:5. [PMID: 30786916 PMCID: PMC6383247 DOI: 10.1186/s40880-019-0351-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 02/14/2019] [Indexed: 12/22/2022] Open
Abstract
Background Glioblastoma (GBM) is a highly virulent tumor of the central nervous system, with a median survival < 15 months. Clearly, an improvement in treatment outcomes is needed. However, the emergence of these malignancies within the delicate brain parenchyma and their infiltrative growth pattern severely limit the use of aggressive local therapies. The particle therapy represents a new promising therapeutic approach to circumvent these prohibitive conditions with improved treatment efficacy. Methods and design Patients with newly diagnosed malignant gliomas will have their tumor tissue samples submitted for the analysis of the status of O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation. In Phase I, the patients will undergo an induction carbon ion radiotherapy (CIRT) boost followed by 60 GyE of proton irradiation with concurrent temozolomide (TMZ) at 75 mg/m2. To determine the maximal dose of safe induction boost, the tolerance, and acute toxicity rates in a dose-escalation manner from 9 to 18 GyE in three fractions will be used. In Phase III, GBM-only patients will be randomized to receive either 60 GyE (2 GyE per fraction) of proton irradiation with concurrent TMZ (control arm) or a CIRT boost (dose determined in Phase I of this trial) followed by 60 GyE of proton irradiation with concurrent TMZ. The primary endpoints are overall survival (OS) and toxicity rates (acute and long-term). Secondary endpoints are progression-free survival (PFS), and tumor response (based upon assessment with C-methionine/fluoro-ethyl-tyrosine positron emission tomography [MET/FET PET] or magnetic resonance imaging [MRI] and detection of serologic immune markers). We hypothesize that the induction CIRT boost will result in a greater initial tumor-killing ability and prime the tumor microenvironment for enhanced immunologic tumor clearance, resulting in an expected 33% improvement in OS rates. Discussion The prognosis of GBM remains grim. The mechanism underpinning the poor prognosis of this malignancy is its chronic state of tumor hypoxia, which promotes both immunosuppression/immunologic evasion and radio-resistance. The unique physical and biological properties of CIRT are expected to overcome these microenvironmental limitations to confer an improved tumor-killing ability and anti-tumor immune response, which could result in an improvement in OS with minimal toxicity. Trial registration number This trial has been registered with the China Clinical Trials Registry, and was allocated the number ChiCTR-OID-17013702.
Collapse
Affiliation(s)
- Lin Kong
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, 201321, P. R. China
| | - Jing Gao
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Pudong, 4365 Kangxin Road, Shanghai, 201321, P. R. China
| | - Jiyi Hu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Pudong, 4365 Kangxin Road, Shanghai, 201321, P. R. China
| | - Rong Lu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Pudong, 4365 Kangxin Road, Shanghai, 201321, P. R. China
| | - Jing Yang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Pudong, 4365 Kangxin Road, Shanghai, 201321, P. R. China
| | - Xianxin Qiu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Pudong, 4365 Kangxin Road, Shanghai, 201321, P. R. China
| | - Weixu Hu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Pudong, 4365 Kangxin Road, Shanghai, 201321, P. R. China
| | - Jiade J Lu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Pudong, 4365 Kangxin Road, Shanghai, 201321, P. R. China.
| |
Collapse
|
44
|
Helm A, Ebner DK, Tinganelli W, Simoniello P, Bisio A, Marchesano V, Durante M, Yamada S, Shimokawa T. Combining Heavy-Ion Therapy with Immunotherapy: An Update on Recent Developments. Int J Part Ther 2018; 5:84-93. [PMID: 31773022 PMCID: PMC6871592 DOI: 10.14338/ijpt-18-00024.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/05/2018] [Indexed: 12/18/2022] Open
Abstract
Clinical trials and case reports of cancer therapies combining radiation therapy with immunotherapy have at times demonstrated total reduction or elimination of metastatic disease. While virtually all trials focus on the use of immunotherapy combined with conventional photon irradiation, the dose-distributive benefits of particles, in particular the distinct biological effects of heavy ions, have unknown potential vis-a-vis systemic disease response. Here, we review recent developments and evidence with a focus on the potential for heavy-ion combination therapy.
Collapse
Affiliation(s)
- Alexander Helm
- Trento Institute for Fundamental Physics and Applications-National Institute for Nuclear Physics, Trento, Italy
| | - Daniel K. Ebner
- Brown University Alpert Medical School, Providence, RI, USA
- Hospital of the National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| | - Walter Tinganelli
- Trento Institute for Fundamental Physics and Applications-National Institute for Nuclear Physics, Trento, Italy
| | - Palma Simoniello
- Department of Science and Technology, Parthenope University of Naples, Naples, Italy
| | - Alessandra Bisio
- Center for Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Valentina Marchesano
- Trento Institute for Fundamental Physics and Applications-National Institute for Nuclear Physics, Trento, Italy
- Center for Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Marco Durante
- Trento Institute for Fundamental Physics and Applications-National Institute for Nuclear Physics, Trento, Italy
| | - Shigeru Yamada
- Hospital of the National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takashi Shimokawa
- National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
45
|
Matsumoto Y, Furusawa Y, Uzawa A, Hirayama R, Koike S, Ando K, Tsuboi K, Sakurai H. Antimetastatic Effects of Carbon-Ion Beams on Malignant Melanomas. Radiat Res 2018; 190:412-423. [DOI: 10.1667/rr15075.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Yoshitaka Matsumoto
- Radiation Oncology, Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, 305-8575, Japan
| | - Yoshiya Furusawa
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Akiko Uzawa
- Department of Charged Particle Therapy Research, Clinical Research Cluster, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Ryoichi Hirayama
- Department of Charged Particle Therapy Research, Clinical Research Cluster, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Sachiko Koike
- Department of Accelerator and Medical Physics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Koichi Ando
- Medicine & Biology Division, Heavy Ion Medical Center, Gunma University, Gunma, 371-8511, Japan
| | - Koji Tsuboi
- Radiation Oncology, Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, 305-8575, Japan
| | - Hideyuki Sakurai
- Radiation Oncology, Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, 305-8575, Japan
| |
Collapse
|
46
|
Tsuboi K. Advantages and Limitations in the Use of Combination Therapies with Charged Particle Radiation Therapy. Int J Part Ther 2018; 5:122-132. [PMID: 31773024 DOI: 10.14338/ijpt-18-00019.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/21/2018] [Indexed: 12/15/2022] Open
Abstract
Purpose Studies are currently underway to help provide basic and clinical evidence for combination particle beam radiation therapy, on which there are few published reports. The purpose of this article is to summarize the current status in the use of particle beams combined with other modalities. Results Following from experiences in x-ray radiation therapy, combination therapy with proton beams (PBT) has been attempted, and several clinical studies have reported improved survival rates for patients with non-small cell lung cancer, pancreatic cancers, esophageal cancers, and glioblastomas. Recently, basic studies combining PBT with PARP inhibitors and histone deacetylase inhibitors have also reported promising results. In the area of carbon ion therapy (CIT), there are few clinical reports on combination therapy; however, the number of basic research reports exceeds that for PBT. So far, the combined use of cytotoxic drugs with CIT yields independent additive effects. In addition, it is notable that combination therapy with CIT is effective against radioresistant cancer stem-like cells. Recent evidence also suggests that local radiation therapy can induce an effective antitumor immune response. There has been an increased use of combination immune-modulating agents and cytokines with particle beams, especially CIT. The field of radiation therapy is evolving from a strong reliance on local-regional treatment to a growing reliance on systemic immunotherapy. Conclusions The combined use of anticancer agents with particle radiation therapy has a considerable potential effect. Future research in molecular targeting therapy and immunotherapy may help identify the most efficacious approach for combination therapy with protons and carbon ions.
Collapse
Affiliation(s)
- Koji Tsuboi
- Proton Medical Research Center, Faculty of Medicine, University of Tsukuba, Japan
| |
Collapse
|
47
|
Lee HJ, Zeng J, Rengan R. Proton beam therapy and immunotherapy: an emerging partnership for immune activation in non-small cell lung cancer. Transl Lung Cancer Res 2018; 7:180-188. [PMID: 29876317 DOI: 10.21037/tlcr.2018.03.28] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Proton beam therapy (PBT) is becoming an increasingly common option for patients undergoing radiation therapy (RT). With the concurrent emergence of immunotherapy as an effective systemic treatment for historically treatment-resistant disease such as advanced non-small cell lung cancer (NSCLC), the combination of RT's immunoadjuvant effects with immunotherapy is gaining widespread attention. However, pre-clinical and clinical studies have shown potential immunosuppressive mechanisms associated with conventional RT that may restrict its immunogenic potential. Protons, as charged particles, exhibit both dosimetric and biological differences in normal and cancer cells that may be able to not only enhance the immunoadjuvant effects of RT, but also reduce immunosuppressive mechanisms. Here, we review the rationale, preclinical and clinical evidence, and ongoing efforts in combining PBT with immunotherapy in cancer treatment with a focus on NSCLC.
Collapse
Affiliation(s)
- Howard J Lee
- University of Washington Medical Center, Seattle, WA, USA
| | - Jing Zeng
- University of Washington Medical Center, Seattle, WA, USA
| | - Ramesh Rengan
- University of Washington Medical Center, Seattle, WA, USA
| |
Collapse
|
48
|
Brownstein JM, Wisdom AJ, Castle KD, Mowery YM, Guida P, Lee CL, Tommasino F, Tessa CL, Scifoni E, Gao J, Luo L, Campos LDS, Ma Y, Williams N, Jung SH, Durante M, Kirsch DG. Characterizing the Potency and Impact of Carbon Ion Therapy in a Primary Mouse Model of Soft Tissue Sarcoma. Mol Cancer Ther 2018; 17:858-868. [PMID: 29437879 PMCID: PMC5912881 DOI: 10.1158/1535-7163.mct-17-0965] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/28/2017] [Accepted: 02/01/2018] [Indexed: 12/11/2022]
Abstract
Carbon ion therapy (CIT) offers several potential advantages for treating cancers compared with X-ray and proton radiotherapy, including increased biological efficacy and more conformal dosimetry. However, CIT potency has not been characterized in primary tumor animal models. Here, we calculate the relative biological effectiveness (RBE) of carbon ions compared with X-rays in an autochthonous mouse model of soft tissue sarcoma. We used Cre/loxP technology to generate primary sarcomas in KrasLSL-G12D/+; p53fl/fl mice. Primary tumors were irradiated with a single fraction of carbon ions (10 Gy), X-rays (20 Gy, 25 Gy, or 30 Gy), or observed as controls. The RBE was calculated by determining the dose of X-rays that resulted in similar time to posttreatment tumor volume quintupling and exponential growth rate as 10 Gy carbon ions. The median tumor volume quintupling time and exponential growth rate of sarcomas treated with 10 Gy carbon ions and 30 Gy X-rays were similar: 27.3 and 28.1 days and 0.060 and 0.059 mm3/day, respectively. Tumors treated with lower doses of X-rays had faster regrowth. Thus, the RBE of carbon ions in this primary tumor model is 3. When isoeffective treatments of carbon ions and X-rays were compared, we observed significant differences in tumor growth kinetics, proliferative indices, and immune infiltrates. We found that carbon ions were three times as potent as X-rays in this aggressive tumor model and identified unanticipated differences in radiation response that may have clinical implications. Mol Cancer Ther; 17(4); 858-68. ©2018 AACR.
Collapse
Affiliation(s)
- Jeremy M Brownstein
- Department of Radiation Oncology, Duke University Health System, Durham, North Carolina
| | - Amy J Wisdom
- Department of Pharmacology & Cancer Biology, Duke University, Durham, North Carolina
| | - Katherine D Castle
- Department of Pharmacology & Cancer Biology, Duke University, Durham, North Carolina
| | - Yvonne M Mowery
- Department of Radiation Oncology, Duke University Health System, Durham, North Carolina
| | - Peter Guida
- Department of Biology, Brookhaven National Laboratory, Upton, New York
| | - Chang-Lung Lee
- Department of Radiation Oncology, Duke University Health System, Durham, North Carolina
| | - Francesco Tommasino
- Trento Institute for Fundamental Physics and Applications, National Institute for Nuclear Physics (INFN), Trento, Italy
- Department of Physics, University of Trento, Trento, Italy
| | - Chiara La Tessa
- Brookhaven National Laboratory, Upton, New York
- Trento Institute for Fundamental Physics and Applications, National Institute for Nuclear Physics (INFN), Trento, Italy
- Department of Physics, University of Trento, Trento, Italy
| | - Emanuele Scifoni
- Trento Institute for Fundamental Physics and Applications, National Institute for Nuclear Physics (INFN), Trento, Italy
| | - Junheng Gao
- Department of Biostatistics and Informatics, Duke University, Durham, North Carolina
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Health System, Durham, North Carolina
| | | | - Yan Ma
- Department of Radiation Oncology, Duke University Health System, Durham, North Carolina
| | - Nerissa Williams
- Department of Radiation Oncology, Duke University Health System, Durham, North Carolina
| | - Sin-Ho Jung
- Department of Biostatistics and Informatics, Duke University, Durham, North Carolina
| | - Marco Durante
- Trento Institute for Fundamental Physics and Applications, National Institute for Nuclear Physics (INFN), Trento, Italy
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Health System, Durham, North Carolina.
- Department of Pharmacology & Cancer Biology, Duke University, Durham, North Carolina
| |
Collapse
|
49
|
Sato K, Nitta N, Aoki I, Imai T, Shimokawa T. Repeated photon and C-ion irradiations in vivo have different impact on alteration of tumor characteristics. Sci Rep 2018; 8:1458. [PMID: 29362374 PMCID: PMC5780469 DOI: 10.1038/s41598-018-19422-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 12/12/2017] [Indexed: 12/16/2022] Open
Abstract
Precise characterization of tumor recurrence and regrowth after radiotherapy are important for prognostic understanding of the therapeutic effect. Here, we established a novel in vivo mouse model for evaluating the characteristics of regrown tumor after repeated photon and carbon ion (C-ion) irradiations. The results showed that tumor growth rate, lung metastasis, shortening of the survival of the tumor-bearing mice, and tumor microvessel formation were promoted 2- to 3-fold, and expression of angiogenic and metastatic genes increased 1.5- to 15-fold in regrown tumors after repeated photon irradiations, whereas repeated C-ion irradiations did not alter these characteristics. Interestingly, both repeated photon and C-ion irradiations did not generate radioresistance, which is generally acquired for in vitro treatment. Our results demonstrated that the repetition of photon, and not C-ion, irradiations in vivo alter the characteristics of the regrown tumor, making it more aggressive without acquisition of radioresistance.
Collapse
Affiliation(s)
- Katsutoshi Sato
- Cancer Metastasis Research Team, Advanced Radiation Biology Research Program, Research Center for Charged Particle Therapy, National Institute of Radiological Sciences, QST, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
- Clinical Genetic Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Koto-Ku, Tokyo, 135-8550, Japan
| | - Nobuhiro Nitta
- Department of Molecular Imaging and Theranostics, and Group of Quantum-state Controlled MRI, National Institute of Radiological Sciences, QST, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Ichio Aoki
- Department of Molecular Imaging and Theranostics, and Group of Quantum-state Controlled MRI, National Institute of Radiological Sciences, QST, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Takashi Imai
- National Institute of Radiological Sciences, QST, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Takashi Shimokawa
- Cancer Metastasis Research Team, Advanced Radiation Biology Research Program, Research Center for Charged Particle Therapy, National Institute of Radiological Sciences, QST, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
| |
Collapse
|
50
|
Kumar SS, Higgins KA, McGarry RC. Emerging Therapies for Stage III Non-Small Cell Lung Cancer: Stereotactic Body Radiation Therapy and Immunotherapy. Front Oncol 2017; 7:197. [PMID: 28929083 PMCID: PMC5591326 DOI: 10.3389/fonc.2017.00197] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 08/17/2017] [Indexed: 12/25/2022] Open
Abstract
The current standard of care for locally advanced non-small cell lung cancer (NSCLC) includes radiation, chemotherapy, and surgery in certain individualized cases. In unresectable NSCLC, chemoradiation has been the standard of care for the past three decades. Local and distant failure remains high in this group of patients, so dose escalation has been studied in both single institution and national clinical trials. Though initial studies showed a benefit to dose escalation, phase III studies examining dose escalation using standard fractionation or hyperfractionation have failed to show a benefit. Over the last 17 years, stereotactic body radiation therapy (SBRT) has shown a high degree of safety and local control for stage I lung cancers and other localized malignancies. More recently, phase I/II studies using SBRT for dose escalation after conventional chemoradiation in locally advanced NSCLC have been promising with good apparent safety. Immunotherapy also offers opportunities to address distant disease and preclinical data suggest immunotherapy in tandem with SBRT may be a rational way to induce an “abscopal effect” although there are little clinical data as yet. By building on the proven concept of conventional chemoradiation for patients with locally advanced NSCLC with a subsequent radiation dose intensification to residual disease with SBRT concurrent with immunotherapy, we hope address the issues of metastatic and local failures. This “quadmodality” approach is still in its infancy but appears to be a safe and rational approach to the improving the outcome of NSCLC therapy.
Collapse
Affiliation(s)
- Sameera S Kumar
- Department of Radiation Medicine, University of Kentucky, Lexington, KY, United States
| | - Kristin A Higgins
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, The Emory Clinic, Atlanta, GA, United States
| | - Ronald C McGarry
- Department of Radiation Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|