1
|
Shao Y, Han S, Hou Z, Yang C, Zhao Y. Tumor-associated macrophages within the immunological milieu: An emerging focal point for therapeutic intervention. Heliyon 2024; 10:e36839. [PMID: 39281573 PMCID: PMC11401039 DOI: 10.1016/j.heliyon.2024.e36839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
Tumor-associated macrophages play an important role in the tumor immune microenvironment, and regulating the function of tumor-associated macrophages has important therapeutic potential in tumor therapy. Mature macrophages could migrate to the tumor microenvironment, influencing multiple factors such as tumor cell proliferation, invasion, metastasis, extracellular matrix remodeling, immune suppression, and drug resistance. As a major component of the tumor microenvironment, tumor-associated macrophages crosstalk with other immune cells. Currently, tumor-associated macrophages have garnered considerable attention in tumor therapy, broadening the spectrum of drug selection to some extent, thereby aiding in mitigating the prevailing clinical drug resistance dilemma. This article summarizes the recent advances in tumor-associated macrophages concerning immunology, drug targeting mechanisms for tumor-associated macrophages treatment, new developments, and existing challenges, offering insights for future therapeutic approaches. In addition, this paper summarized the impact of tumor-associated macrophages on current clinical therapies, discussed the advantages and disadvantages of targeted tumor-associated macrophages therapy compared with existing tumor therapies, and predicted and discussed the future role of targeted tumor-associated macrophages therapy and the issues that need to be focused on.
Collapse
Affiliation(s)
- Yanchi Shao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Song Han
- The First Hospital of Jilin University, Changchun, China
| | - Zhenxin Hou
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Chen Yang
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yanbin Zhao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
2
|
Zhou L, Liu Y, Wu Y, Yang X, Spring Kong FM, Lu Y, Xue J. Low-dose radiation therapy mobilizes antitumor immunity: New findings and future perspectives. Int J Cancer 2024; 154:1143-1157. [PMID: 38059788 DOI: 10.1002/ijc.34801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023]
Abstract
Radiotherapy has unique immunostimulatory and immunosuppressive effects. Although high-dose radiotherapy has been found to have systemic antitumor effects, clinically significant abscopal effects were uncommon on the basis of irradiating single lesion. Low-dose radiation therapy (LDRT) emerges as a novel approach to enhance the antitumor immune response due to its role as a leverage to reshape the tumor immune microenvironment (TIME). In this article, from bench to bedside, we reviewed the possible immunomodulatory role of LDRT on TIME and systemic tumor immune environment, and outlined preclinical evidence and clinical application. We also discussed the current challenges when LDRT is used as a combination therapy, including the optimal dose, fraction, frequency, and combination of drugs. The advantage of low toxicity makes LDRT potential to be applied in multiple lesions to amplify antitumor immune response in polymetastatic disease, and its intersection with other disciplines might also make it a direction for radiotherapy-combined modalities.
Collapse
Affiliation(s)
- Laiyan Zhou
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Disaster Medical Center, Sichuan University, Chengdu, China
| | - Yuanxin Liu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanjun Wu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Yang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feng-Ming Spring Kong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Cornice J, Verzella D, Arboretto P, Vecchiotti D, Capece D, Zazzeroni F, Franzoso G. NF-κB: Governing Macrophages in Cancer. Genes (Basel) 2024; 15:197. [PMID: 38397187 PMCID: PMC10888451 DOI: 10.3390/genes15020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are the major component of the tumor microenvironment (TME), where they sustain tumor progression and or-tumor immunity. Due to their plasticity, macrophages can exhibit anti- or pro-tumor functions through the expression of different gene sets leading to distinct macrophage phenotypes: M1-like or pro-inflammatory and M2-like or anti-inflammatory. NF-κB transcription factors are central regulators of TAMs in cancers, where they often drive macrophage polarization toward an M2-like phenotype. Therefore, the NF-κB pathway is an attractive therapeutic target for cancer immunotherapy in a wide range of human tumors. Hence, targeting NF-κB pathway in the myeloid compartment is a potential clinical strategy to overcome microenvironment-induced immunosuppression and increase anti-tumor immunity. In this review, we discuss the role of NF-κB as a key driver of macrophage functions in tumors as well as the principal strategies to overcome tumor immunosuppression by targeting the NF-κB pathway.
Collapse
Affiliation(s)
- Jessica Cornice
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Paola Arboretto
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| | - Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Guido Franzoso
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| |
Collapse
|
4
|
Au NPB, Wu T, Kumar G, Jin Y, Li YYT, Chan SL, Lai JHC, Chan KWY, Yu KN, Wang X, Ma CHE. Low-dose ionizing radiation promotes motor recovery and brain rewiring by resolving inflammatory response after brain injury and stroke. Brain Behav Immun 2024; 115:43-63. [PMID: 37774892 DOI: 10.1016/j.bbi.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/24/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023] Open
Abstract
Traumatic brain injury (TBI) and stroke share a common pathophysiology that worsens over time due to secondary tissue injury caused by sustained inflammatory response. However, studies on pharmacological interventions targeting the complex secondary injury cascade have failed to show efficacy. Here, we demonstrated that low-dose ionizing radiation (LDIR) reduced lesion size and reversed motor deficits after TBI and photothrombotic stroke. Magnetic resonance imaging demonstrated significant reduction of infarct volume in LDIR-treated mice after stroke. Systems-level transcriptomic analysis showed that genes upregulated in LDIR-treated stoke mice were enriched in pathways associated with inflammatory and immune response involving microglia. LDIR induced upregulation of anti-inflammatory- and phagocytosis-related genes, and downregulation of key pro-inflammatory cytokine production. These findings were validated by live-cell assays, in which microglia exhibited higher chemotactic and phagocytic capacities after LDIR. We observed substantial microglial clustering at the injury site, glial scar clearance and reversal of motor deficits after stroke. Cortical microglia/macrophages depletion completely abolished the beneficial effect of LDIR on motor function recovery in stroke mice. LDIR promoted axonal projections (brain rewiring) in motor cortex and recovery of brain activity detected by electroencephalography recordings months after stroke. LDIR treatment delayed by 8 h post-injury still maintained full therapeutic effects on motor recovery, indicating that LDIR is a promising therapeutic strategy for TBI and stroke.
Collapse
Affiliation(s)
| | - Tan Wu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Department of Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Yuting Jin
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | | | - Shun Lam Chan
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Joseph Ho Chi Lai
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Kannie Wai Yan Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Kwan Ngok Yu
- Department of Physics, City University of Hong Kong, Hong Kong, China
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Department of Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Chi Him Eddie Ma
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
5
|
Zhao F, Tian H, Wang Y, Zhang J, Liu F, Fu L. LINC01004-SPI1 axis-activated SIGLEC9 in tumor-associated macrophages induces radioresistance and the formation of immunosuppressive tumor microenvironment in esophageal squamous cell carcinoma. Cancer Immunol Immunother 2023; 72:1835-1851. [PMID: 36688997 DOI: 10.1007/s00262-022-03364-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/27/2022] [Indexed: 01/24/2023]
Abstract
Radioresistance and immunosuppression remain the major obstacles in the anti-cancer treatments. This work studies the functions of sialic acid binding Ig like lectin 9 (SIGLEC9) and its related molecules in radioresistance and immunosuppression in esophageal squamous cell carcinoma (ESCC). The single-cell analysis showed that SIGLEC9 was mainly expressed on tumor-associated macrophages (TAMs). Monocytes-derived macrophages were co-cultured with ESCC cells and subjected to radiotherapy. High or low doses of radiotherapy induced SIGLEC9 upregulation and M2 polarization of TAMs. Artificial inhibition of SIGLEC9 in TAMs suppressed the radioresistance and immunosuppressive tumor microenvironment (TME) in the co-cultured ESCC cells. Upstream molecules of SIGLEC9 were predicted via bioinformatics. LINC01004 recruited Spi-1 proto-oncogene (SPI1) in nucleus of TAMs to induce transcriptional activation of SIGLEC9. SIGLEC9 interacted with mucin 1 (MUC1). MUC1 overexpression in ESCCs induced M2 skewing of TAMs, enhanced radioresistance and immunosuppression, and promoted nuclear translocation of β-catenin to suppress radiotherapy-induced ferroptosis of ESCC cells. These effects were blocked upon SIGLEC9 suppression. In vitro results were reproduced in the animal models with xenograft tumors. Taken together, this study demonstrates that the LINC01004-SPI1 axis-activated SIGLEC9 in TAMs induces radioresistance and the formation of immunosuppressive TME in ESCC.
Collapse
Affiliation(s)
- Fen Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, 250117, Shandong, People's Republic of China
| | - Hui Tian
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Yungang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China
| | - Jianbo Zhang
- Departments of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China
| | - Fang Liu
- Department of Imaging, Shandong Medical College, Jinan, 250002, Shandong, People's Republic of China
| | - Lei Fu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China.
| |
Collapse
|
6
|
Deloch L, Rückert M, Weissmann T, Lettmaier S, Titova E, Wolff T, Weinrich F, Fietkau R, Gaipl US. The various functions and phenotypes of macrophages are also reflected in their responses to irradiation: A current overview. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 376:99-120. [PMID: 36997271 DOI: 10.1016/bs.ircmb.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Macrophages are a vital part of the innate immune system that are involved in healthy biological processes but also in disease modulation and response to therapy. Ionizing radiation is commonly used in the treatment of cancer and, in a lower dose range, as additive therapy for inflammatory diseases. In general, lower doses of ionizing radiation are known to induce rather anti-inflammatory responses, while higher doses are utilized in cancer treatment where they result, next to tumor control, in rather inflammatory responses. Most experiments that have been carried out in ex vivo on macrophages find this to be true, however in vivo, tumor-associated macrophages, for example, show a contradictory response to the respective dose-range. While some knowledge in radiation-induced modulations of macrophages has been collected, many of the underlying mechanisms remain unclear. Due to their pivotal role in the human body, however, they are a great target in therapy and could potentially aid in better treatment outcome. We therefore summarized the current knowledge of macrophage mediated radiation responses.
Collapse
|
7
|
Pinto AT, Machado AB, Osório H, Pinto ML, Vitorino R, Justino G, Santa C, Castro F, Cruz T, Rodrigues C, Lima J, Sousa JLR, Cardoso AP, Figueira R, Monteiro A, Marques M, Manadas B, Pauwels J, Gevaert K, Mareel M, Rocha S, Duarte T, Oliveira MJ. Macrophage Resistance to Ionizing Radiation Exposure Is Accompanied by Decreased Cathepsin D and Increased Transferrin Receptor 1 Expression. Cancers (Basel) 2022; 15:270. [PMID: 36612268 PMCID: PMC9818572 DOI: 10.3390/cancers15010270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/06/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
PURPOSE To identify a molecular signature of macrophages exposed to clinically relevant ionizing radiation (IR) doses, mirroring radiotherapy sessions. METHODS Human monocyte-derived macrophages were exposed to 2 Gy/ fraction/ day for 5 days, mimicking one week of cancer patient's radiotherapy. Protein expression profile by proteomics was performed. RESULTS A gene ontology analysis revealed that radiation-induced protein changes are associated with metabolic alterations, which were further supported by a reduction of both cellular ATP levels and glucose uptake. Most of the radiation-induced deregulated targets exhibited a decreased expression, as was the case of cathepsin D, a lysosomal protease associated with cell death, which was validated by Western blot. We also found that irradiated macrophages exhibited an increased expression of the transferrin receptor 1 (TfR1), which is responsible for the uptake of transferrin-bound iron. TfR1 upregulation was also found in tumor-associated mouse macrophages upon tumor irradiation. In vitro irradiated macrophages also presented a trend for increased divalent metal transporter 1 (DMT1), which transports iron from the endosome to the cytosol, and a significant increase in iron release. CONCLUSIONS Irradiated macrophages present lower ATP levels and glucose uptake, and exhibit decreased cathepsin D expression, while increasing TfR1 expression and altering iron metabolism.
Collapse
Affiliation(s)
- Ana Teresa Pinto
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), Universidade de Aveiro, 3810-193 Aveiro, Portugal
| | - Ana Beatriz Machado
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Champalimaud Centre for the Unknown, Fundação Champalimaud, 1400-038 Lisboa, Portugal
| | - Hugo Osório
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP–Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal
- Departament of Pathology, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Marta Laranjeiro Pinto
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Rui Vitorino
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), Universidade de Aveiro, 3810-193 Aveiro, Portugal
| | - Gonçalo Justino
- Centro de Química Estrutural–Institute of Molecular Sciences, Instituto Superior Técnico, Universidade Técnica de Lisboa, 1049-001 Lisboa, Portugal
| | - Cátia Santa
- CNC–Center for Neuroscience and Cell Biology, Universidade de Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), Universidade de Coimbra, 3030-789 Coimbra, Portugal
| | - Flávia Castro
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Tânia Cruz
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Carla Rodrigues
- REQUIMTE–LAQV, Chemistry Department, NOVA School of Science and Technology, Universidade de Lisboa, 2829-516 Caparica, Portugal
| | - Jorge Lima
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP–Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal
| | - José Luís R. Sousa
- Personal Health Data Science Group, Sano-Centre for Computational Personalised Medicine, 30-054 Krakow, Poland
| | - Ana Patrícia Cardoso
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Rita Figueira
- Radiotherapy Service, Centro Hospitalar Universitário São João (CHUSJ), EPE, 4200-319 Porto, Portugal
| | - Armanda Monteiro
- Radiotherapy Service, Centro Hospitalar Universitário São João (CHUSJ), EPE, 4200-319 Porto, Portugal
| | - Margarida Marques
- Radiotherapy Service, Centro Hospitalar Universitário São João (CHUSJ), EPE, 4200-319 Porto, Portugal
| | - Bruno Manadas
- Institute for Interdisciplinary Research (III), Universidade de Coimbra, 3030-789 Coimbra, Portugal
| | - Jarne Pauwels
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Kris Gevaert
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Marc Mareel
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, 9000 Ghent, Belgium
| | - Sónia Rocha
- Institute of System, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3 GE, UK
| | - Tiago Duarte
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria José Oliveira
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Departament of Pathology, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| |
Collapse
|
8
|
Concerted regulation of OPG/RANKL/ NF‑κB/MMP-13 trajectories contribute to ameliorative capability of prodigiosin and/or low dose γ-radiation against adjuvant- induced arthritis in rats. Int Immunopharmacol 2022; 111:109068. [PMID: 35944459 DOI: 10.1016/j.intimp.2022.109068] [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: 05/27/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prodigiosin (PDG) is a microbial red dye with antioxidant and anti-inflammatory properties, although its effect on rheumatoid arthritis (RA) remains uncertain. Also, multiple doses of low dose γ- radiation (LDR) have been observed to be as a successful intervention for RA. Thus, the purpose of this study was to investigate the ameliorative potential of PDG and/or LDR on adjuvant-induced arthritis (AIA) in rats. METHODS The anti-inflammatory and anti-arthritic effects of PDG and/or LDR were examined in vitro and in vivo, respectively. In the AIA model, the arthritic indexes, paw swelling degrees, body weight gain, and histopathological assessment in AIA rats were assayed. The impact of PDG (200 µg/kg; p.o) and/or LDR (0.5 Gy) on the levels of pro- and anti-inflammatory cytokines (IL-1β, TNF-α, IL-6, IL-18, IL-17A, and IL-10) as well as the regulation of osteoprotegrin (OPG)/ receptor activator of nuclear factor κB ligand (RANKL)/ nuclear factor-κB (NF-κB)/MMP-13 pathways was determined. Methotrexate (MTX; 0.05 mg/kg; twice/week, i.p) was administered concurrently as a standard anti-arthritic drug. RESULTS PDG and/or LDR markedly diminished the arthritic indexes, paw edema, weigh loss in AIA rats, alleviated the pathological alterations in joints, reduced the levels of pro-inflammatory cytokines IL-1β, TNF-α, IL-6, IL-18, IL-17A, and RANKL in serum and synovial tissues, while increasing anti-inflammatory cytokines IL-10 and OPG levels. Moreover, PDG and/or LDR down-regulated the expression of RANKL, NF-κBp65, MMP13, caspase-3, and decreased the RANKL/OPG ratio, whereas OPG and collagen II were enhanced in synovial tissues. CONCLUSION PDG and/or LDR exhibited obvious anti-RA activity on AIA.
Collapse
|
9
|
Rodríguez-Tomàs E, Acosta JC, Torres-Royo L, De Febrer G, Baiges-Gaya G, Castañé H, Jiménez A, Vasco C, Araguas P, Gómez J, Malave B, Árquez M, Calderón D, Piqué B, Algara M, Montero Á, Simó JM, Gabaldó-Barrios X, Sabater S, Camps J, Joven J, Arenas M. Effect of Low-Dose Radiotherapy on the Circulating Levels of Paraoxonase-1-Related Variables and Markers of Inflammation in Patients with COVID-19 Pneumonia. Antioxidants (Basel) 2022; 11:antiox11061184. [PMID: 35740079 PMCID: PMC9220239 DOI: 10.3390/antiox11061184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/04/2022] [Accepted: 06/10/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of our study was to investigate the changes produced by low-dose radiotherapy (LDRT) in the circulating levels of the antioxidant enzyme paraoxonase-1 (PON1) and inflammatory markers in patients with COVID-19 pneumonia treated with LDRT and their interactions with clinical and radiological changes. Data were collected from the IPACOVID prospective clinical trial (NCT04380818). The study included 30 patients treated with a whole-lung dose of 0.5 Gy. Clinical follow-up, as well as PON1-related variables, cytokines, and radiological parameters were analyzed before LDRT, at 24 h, and 1 week after treatment. Twenty-five patients (83.3%) survived 1 week after LDRT. Respiratory function and radiological images improved in survivors. Twenty-four hours after LDRT, PON1 concentration significantly decreased, while transforming growth factor beta 1 (TGF-β1) increased with respect to baseline. One week after LDRT, patients had increased PON1 activities and lower PON1 and TGF-β1 concentrations compared with 24 h after LDRT, PON1 specific activity increased, lactate dehydrogenase (LDH), and C-reactive protein (CRP) decreased, and CD4+ and CD8+ cells increased after one week. Our results highlight the benefit of LDRT in patients with COVID-19 pneumonia and it might be mediated, at least in part, by an increase in serum PON1 activity at one week and an increase in TGF-β1 concentrations at 24 h.
Collapse
Affiliation(s)
- Elisabet Rodríguez-Tomàs
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Tarragona, Spain; (E.R.-T.); (J.C.A.); (L.T.-R.); (P.A.); (J.G.); (B.M.); (M.Á.); (D.C.); (B.P.)
- Unitat de Recerca Biomèdica, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43003 Tarragona, Spain; (G.B.-G.); (H.C.); (A.J.); (J.C.); (J.J.)
| | - Johana C. Acosta
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Tarragona, Spain; (E.R.-T.); (J.C.A.); (L.T.-R.); (P.A.); (J.G.); (B.M.); (M.Á.); (D.C.); (B.P.)
| | - Laura Torres-Royo
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Tarragona, Spain; (E.R.-T.); (J.C.A.); (L.T.-R.); (P.A.); (J.G.); (B.M.); (M.Á.); (D.C.); (B.P.)
| | - Gabriel De Febrer
- Department of Geriatric and Palliative Care, Hospital Universitari Sant Joan de Reus, 43204 Tarragona, Spain; (G.D.F.); (C.V.)
| | - Gerard Baiges-Gaya
- Unitat de Recerca Biomèdica, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43003 Tarragona, Spain; (G.B.-G.); (H.C.); (A.J.); (J.C.); (J.J.)
| | - Helena Castañé
- Unitat de Recerca Biomèdica, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43003 Tarragona, Spain; (G.B.-G.); (H.C.); (A.J.); (J.C.); (J.J.)
| | - Andrea Jiménez
- Unitat de Recerca Biomèdica, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43003 Tarragona, Spain; (G.B.-G.); (H.C.); (A.J.); (J.C.); (J.J.)
| | - Carlos Vasco
- Department of Geriatric and Palliative Care, Hospital Universitari Sant Joan de Reus, 43204 Tarragona, Spain; (G.D.F.); (C.V.)
| | - Pablo Araguas
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Tarragona, Spain; (E.R.-T.); (J.C.A.); (L.T.-R.); (P.A.); (J.G.); (B.M.); (M.Á.); (D.C.); (B.P.)
| | - Junior Gómez
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Tarragona, Spain; (E.R.-T.); (J.C.A.); (L.T.-R.); (P.A.); (J.G.); (B.M.); (M.Á.); (D.C.); (B.P.)
| | - Bárbara Malave
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Tarragona, Spain; (E.R.-T.); (J.C.A.); (L.T.-R.); (P.A.); (J.G.); (B.M.); (M.Á.); (D.C.); (B.P.)
| | - Miguel Árquez
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Tarragona, Spain; (E.R.-T.); (J.C.A.); (L.T.-R.); (P.A.); (J.G.); (B.M.); (M.Á.); (D.C.); (B.P.)
| | - David Calderón
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Tarragona, Spain; (E.R.-T.); (J.C.A.); (L.T.-R.); (P.A.); (J.G.); (B.M.); (M.Á.); (D.C.); (B.P.)
| | - Berta Piqué
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Tarragona, Spain; (E.R.-T.); (J.C.A.); (L.T.-R.); (P.A.); (J.G.); (B.M.); (M.Á.); (D.C.); (B.P.)
- Department of Pathology, Hospital Universitari Sant Joan de Reus, 43204 Tarragona, Spain
| | - Manel Algara
- Department of Radiation Oncology, Institut d’Investigacions Mèdiques, Hospital del Mar, Autonomous University of Barcelona, 08193 Barcelona, Spain;
| | - Ángel Montero
- Department of Radiation Oncology, HM Hospitales, 28050 Madrid, Spain;
| | - Josep M. Simó
- Laboratori de Referència Sud, Hospital Universitari Sant Joan de Reus, 43204 Tarragona, Spain; (J.M.S.); (X.G.-B.)
| | - Xavier Gabaldó-Barrios
- Laboratori de Referència Sud, Hospital Universitari Sant Joan de Reus, 43204 Tarragona, Spain; (J.M.S.); (X.G.-B.)
| | - Sebastià Sabater
- Department of Radiation Oncology, Complejo Hospitalario de Albacete, 02006 Albacete, Spain;
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43003 Tarragona, Spain; (G.B.-G.); (H.C.); (A.J.); (J.C.); (J.J.)
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43003 Tarragona, Spain; (G.B.-G.); (H.C.); (A.J.); (J.C.); (J.J.)
| | - Meritxell Arenas
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Tarragona, Spain; (E.R.-T.); (J.C.A.); (L.T.-R.); (P.A.); (J.G.); (B.M.); (M.Á.); (D.C.); (B.P.)
- Correspondence:
| |
Collapse
|
10
|
Cao X, Lai SWT, Chen S, Wang S, Feng M. Targeting tumor-associated macrophages for cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 368:61-108. [PMID: 35636930 DOI: 10.1016/bs.ircmb.2022.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Tumor-associated macrophages (TAMs) are one of the most abundant immune components in the tumor microenvironment and play a plethora of roles in regulating tumorigenesis. Therefore, the therapeutic targeting of TAMs has emerged as a new paradigm for immunotherapy of cancer. Herein, the review summarizes the origin, polarization, and function of TAMs in the progression of malignant diseases. The understanding of such knowledge leads to several distinct therapeutic strategies to manipulate TAMs to battle cancer, which include those to reduce TAM abundance, such as depleting TAMs or inhibiting their recruitment and differentiation, and those to harness or boost the anti-tumor activities of TAMs such as blocking phagocytosis checkpoints, inducing antibody-dependent cellular phagocytosis, and reprogramming TAM polarization. In addition, modulation of TAMs may reshape the tumor microenvironment and therefore synergize with other cancer therapeutics. Therefore, the rational combination of TAM-targeting therapeutics with conventional therapies including radiotherapy, chemotherapy, and other immunotherapies is also reviewed. Overall, targeting TAMs presents itself as a promising strategy to add to the growing repertoire of treatment approaches in the fight against cancer, and it is hopeful that these approaches currently being pioneered will serve to vastly improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| | - Seigmund W T Lai
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Siqi Chen
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Sadira Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| |
Collapse
|
11
|
Dove AP, Cmelak A, Darrow K, McComas KN, Chowdhary M, Beckta J, Kirschner AN. The Use of Low-Dose Radiotherapy in Osteoarthritis: A Review. Int J Radiat Oncol Biol Phys 2022; 114:203-220. [DOI: 10.1016/j.ijrobp.2022.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/08/2022] [Accepted: 04/21/2022] [Indexed: 11/26/2022]
|
12
|
Macaeva E, Tabury K, Michaux A, Janssen A, Averbeck N, Moreels M, De Vos WH, Baatout S, Quintens R. High-LET Carbon and Iron Ions Elicit a Prolonged and Amplified p53 Signaling and Inflammatory Response Compared to low-LET X-Rays in Human Peripheral Blood Mononuclear Cells. Front Oncol 2021; 11:768493. [PMID: 34888245 PMCID: PMC8649625 DOI: 10.3389/fonc.2021.768493] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/01/2021] [Indexed: 12/29/2022] Open
Abstract
Understanding the differences in biological response to photon and particle radiation is important for optimal exploitation of particle therapy for cancer patients, as well as for the adequate application of radiation protection measures for astronauts. To address this need, we compared the transcriptional profiles of isolated peripheral blood mononuclear cells 8 h after exposure to 1 Gy of X-rays, carbon ions or iron ions with those of non-irradiated cells using microarray technology. All genes that were found differentially expressed in response to either radiation type were up-regulated and predominantly controlled by p53. Quantitative PCR of selected genes revealed a significantly higher up-regulation 24 h after exposure to heavy ions as compared to X-rays, indicating their prolonged activation. This coincided with increased residual DNA damage as evidenced by quantitative γH2AX foci analysis. Furthermore, despite the converging p53 signature between radiation types, specific gene sets related to the immune response were significantly enriched in up-regulated genes following irradiation with heavy ions. In addition, irradiation, and in particular exposure to carbon ions, promoted transcript variation. Differences in basal and iron ion exposure-induced expression of DNA repair genes allowed the identification of a donor with distinct DNA repair profile. This suggests that gene signatures may serve as a sensitive indicator of individual DNA damage repair capacity. In conclusion, we have shown that photon and particle irradiation induce similar transcriptional pathways, albeit with variable amplitude and timing, but also elicit radiation type-specific responses that may have implications for cancer progression and treatment
Collapse
Affiliation(s)
- Ellina Macaeva
- Radiobiology Unit, Studiecentrum voor kernenergie - Centre d'étude de l'énergie nucléaire (SCK CEN), Mol, Belgium.,Department of Molecular Biotechnology, Ghent University, Ghent, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Kevin Tabury
- Radiobiology Unit, Studiecentrum voor kernenergie - Centre d'étude de l'énergie nucléaire (SCK CEN), Mol, Belgium.,Department of Biomedical Engineering, University of South Carolina, Columbia, SC, United States
| | - Arlette Michaux
- Radiobiology Unit, Studiecentrum voor kernenergie - Centre d'étude de l'énergie nucléaire (SCK CEN), Mol, Belgium
| | - Ann Janssen
- Radiobiology Unit, Studiecentrum voor kernenergie - Centre d'étude de l'énergie nucléaire (SCK CEN), Mol, Belgium
| | - Nicole Averbeck
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Marjan Moreels
- Radiobiology Unit, Studiecentrum voor kernenergie - Centre d'étude de l'énergie nucléaire (SCK CEN), Mol, Belgium
| | - Winnok H De Vos
- Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Studiecentrum voor kernenergie - Centre d'étude de l'énergie nucléaire (SCK CEN), Mol, Belgium.,Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - Roel Quintens
- Radiobiology Unit, Studiecentrum voor kernenergie - Centre d'étude de l'énergie nucléaire (SCK CEN), Mol, Belgium
| |
Collapse
|
13
|
Abdel-Aziz N, Elkady AA, Elgazzar EM. Effect of Low-Dose Gamma Radiation and Lipoic Acid on High- Radiation-Dose Induced Rat Brain Injuries. Dose Response 2021; 19:15593258211044845. [PMID: 34759786 PMCID: PMC8573698 DOI: 10.1177/15593258211044845] [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: 07/17/2021] [Accepted: 08/17/2021] [Indexed: 12/04/2022] Open
Abstract
Aim This work aims to investigate the possible radio-adaptive mechanisms induced by low-dose (LD) whole-body γ-irradiation alone or combined with alpha-lipoic acid (ALA) administration in modulating high-dose (HD) head irradiation–induced brain injury in rats. Materials and Methods Rats were irradiated with LD (.25 Gy) 24 hours prior HD (20 Gy), and subjected to ALA (100 mg/kg/day) 5 minutes after HD and continued for 10 days. At the end of the experiment, animals were sacrificed and brain samples were dissected for biochemical and histopathological examinations. Results HD irradiation-induced brain injury as manifested by elevation of oxidative stress, DNA damage, apoptotic, and inflammatory markers in brain tissue. Histological examination of brain sections showed marked alterations. However, LD alone or combined with ALA ameliorated the changes induced by HD. Conclusion Under the present experimental conditions, LD whole-body irradiation exhibited neuroprotective activity against detrimental effects of a subsequent HD head irradiation. This effect might be due to the adaptive response induced by LD that activated the anti-oxidative, anti-apoptotic, and anti-inflammatory mechanisms in the affected animals making them able to cope with the subsequent high-dose exposure. However, the combined LD exposure and ALA supplementation produced a further modulating effect in the HD-irradiated rats.
Collapse
Affiliation(s)
- Nahed Abdel-Aziz
- Radiation Biology Research Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Ahmed A Elkady
- Ahmed A. Elkady: Health Radiation Research Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Eman M Elgazzar
- Ahmed A. Elkady: Health Radiation Research Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| |
Collapse
|
14
|
Arenas M, Algara M, De Febrer G, Rubio C, Sanz X, de la Casa MA, Vasco C, Marín J, Fernández-Letón P, Villar J, Torres-Royo L, Villares P, Membrive I, Acosta J, López-Cano M, Araguas P, Quera J, Rodríguez-Tomás F, Montero A. Could pulmonary low-dose radiation therapy be an alternative treatment for patients with COVID-19 pneumonia? Preliminary results of a multicenter SEOR-GICOR nonrandomized prospective trial (IPACOVID trial). Strahlenther Onkol 2021; 197:1010-1020. [PMID: 34230996 PMCID: PMC8260020 DOI: 10.1007/s00066-021-01803-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/01/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE To evaluate the efficacy and safety of lung low-dose radiation therapy (LD-RT) for pneumonia in patients with coronavirus disease 2019 (COVID-19). MATERIALS AND METHODS Inclusion criteria comprised patients with COVID-19-related moderate-severe pneumonia warranting hospitalization with supplemental O2 and not candidates for admission to the intensive care unit because of comorbidities or general status. All patients received single lung dose of 0.5 Gy. Respiratory and systemic inflammatory parameters were evaluated before irradiation, at 24 h and 1 week after LD-RT. Primary endpoint was increased in the ratio of arterial oxygen partial pressure (PaO2) or the pulse oximetry saturation (SpO2) to fractional inspired oxygen (FiO2) ratio of at least 20% at 24 h with respect to the preirradiation value. RESULTS Between June and November 2020, 36 patients with COVID-19 pneumonia and a mean age of 84 years were enrolled. Seventeen were women and 19 were men and all of them had comorbidities. All patients had bilateral pulmonary infiltrates on chest X‑ray. All patients received dexamethasone treatment. Mean SpO2 pretreatment value was 94.28% and the SpO2/FiO2 ratio varied from 255 mm Hg to 283 mm Hg at 24 h and to 381 mm Hg at 1 week, respectively. In those who survived (23/36, 64%), a significant improvement was observed in the percentage of lung involvement in the CT scan at 1 week after LD-RT. No adverse effects related to radiation treatment have been reported. CONCLUSIONS LD-RT appears to be a feasible and safe option in a population with COVID-19 bilateral interstitial pneumonia in the presence of significant comorbidities.
Collapse
Affiliation(s)
- M. Arenas
- Universitat Rovira i Virgili, Tarragona, Spain
- Institut d’Investigacions Pere Virgili, Tarragona, Spain
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Tarragona, Spain
| | - M. Algara
- Department of Radiation Oncology, Hospital del Mar, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
- Institut Hospital del Mar d’Investigacions Mèdiques, Barcelona, Spain
| | - G. De Febrer
- Universitat Rovira i Virgili, Tarragona, Spain
- Department of Geriatric and Palliative care, Hospital Universitari Sant Joan de Reus, Tarragona, Spain
| | - C. Rubio
- Department of Radiation Oncology, HM Hospitales., Madrid, Spain
| | - X. Sanz
- Department of Radiation Oncology, Hospital del Mar, Barcelona, Spain
- Institut Hospital del Mar d’Investigacions Mèdiques, Barcelona, Spain
- Pompeu Fabra University Barcelona, Barcelona, Spain
| | | | - C. Vasco
- Universitat Rovira i Virgili, Tarragona, Spain
- Department of Geriatric and Palliative care, Hospital Universitari Sant Joan de Reus, Tarragona, Spain
| | - J. Marín
- Institut Hospital del Mar d’Investigacions Mèdiques, Barcelona, Spain
- Department of Critical Care, Hospital del Mar, Barcelona, Spain
| | | | - J. Villar
- Institut Hospital del Mar d’Investigacions Mèdiques, Barcelona, Spain
- Department of Infection Diseases, Hospital del Mar, Barcelona, Spain
| | - L. Torres-Royo
- Universitat Rovira i Virgili, Tarragona, Spain
- Institut d’Investigacions Pere Virgili, Tarragona, Spain
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Tarragona, Spain
| | - P. Villares
- Department of Internal Medicine, HM Hospitales, Madrid, Spain
| | - I. Membrive
- Department of Radiation Oncology, Hospital del Mar, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - J. Acosta
- Universitat Rovira i Virgili, Tarragona, Spain
- Institut d’Investigacions Pere Virgili, Tarragona, Spain
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Tarragona, Spain
| | - M. López-Cano
- Department of Internal Medicine, HM Hospitales, Madrid, Spain
| | - P. Araguas
- Universitat Rovira i Virgili, Tarragona, Spain
- Institut d’Investigacions Pere Virgili, Tarragona, Spain
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Tarragona, Spain
| | - J. Quera
- Department of Radiation Oncology, Hospital del Mar, Barcelona, Spain
- Institut Hospital del Mar d’Investigacions Mèdiques, Barcelona, Spain
- Pompeu Fabra University Barcelona, Barcelona, Spain
| | - F. Rodríguez-Tomás
- Universitat Rovira i Virgili, Tarragona, Spain
- Institut d’Investigacions Pere Virgili, Tarragona, Spain
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Tarragona, Spain
| | - A. Montero
- Department of Radiation Oncology, HM Hospitales., Madrid, Spain
| |
Collapse
|
15
|
Álvarez B, Montero A, Alonso R, Valero J, López M, Ciérvide R, Sánchez E, Hernando O, García-Aranda M, Martí J, Prado A, Chen-Zhao X, Rubio C. Low-dose radiation therapy for hand osteoarthritis: shaking hands again? Clin Transl Oncol 2021; 24:532-539. [PMID: 34585316 DOI: 10.1007/s12094-021-02710-w] [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: 07/02/2021] [Accepted: 09/13/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Hand osteoarthritis (HOA) is one of the most common causes of pain and functional disability in western countries and there is still no definitive cure. Low-dose radiation therapy (LDRT) has anti-inflammatory properties that have shown to be effective in the symptomatic relief of various degenerative musculoskeletal disorders. We designed a clinical protocol using LDRT for symptomatic HOA and present results and tolerance in the first 100 patients included. MATERIALS AND METHODS Between April 2015 and March 2021, 100 patients with a median age of 60 were treated. Fifty-seven patients suffering from proximal/distal interphalangeal joint pain, 40 patients with thumb arthritis, 2 patients with radiocarpal joint affection and 1 patient with metacarpophalangeal joint pain were enrolled. LDRT comprised of 6 fractions of 0.5-1 Gy on every other day up to a total dose of 3-6 Gy. Clinical response was evaluated according to the visual analog scale (VAS) for pain level and the von Pannewitz score (VPS) for joint functionality. Any patients not achieving subjective adequate pain relief after 8 weeks of treatment were offered a second identical LDRT course. RESULTS With a median follow-up of 10.5 months (range 7.55-12.45), 94% reported an improvement in the pain, with a significant reduction in the VAS level after 3, 6 and 12 months (p < 0.001). Sixty-three patients needed a second course of treatment at a median time interval of 12 weeks (range 9-14). The mean VAS score before treatment was 8 (range 3-10). After treatment, it was 5 (range 1-10). After 3, 6 and 12 months, the mean VAS scores were 4 (range 0-9), 3 (range 0-9) and 3.5 (range 0-9), respectively. Seventy patients reported functionality improvements after LDRT according to the von Pannewitz score. No acute or late complications were observed. CONCLUSION LDRT appears to be safe and useful for HOA and is associated with good rates of pain relief and functionality improvements. However, further studies are necessary to confirm these promising results.
Collapse
Affiliation(s)
- B Álvarez
- Department of Radiation Oncology, HM Hospitales, Calle Oña 10, 28050, Madrid, Spain
| | - A Montero
- Department of Radiation Oncology, HM Hospitales, Calle Oña 10, 28050, Madrid, Spain.
| | - R Alonso
- Department of Radiation Oncology, HM Hospitales, Calle Oña 10, 28050, Madrid, Spain
| | - J Valero
- Department of Radiation Oncology, HM Hospitales, Calle Oña 10, 28050, Madrid, Spain
| | - M López
- Department of Radiation Oncology, HM Hospitales, Calle Oña 10, 28050, Madrid, Spain
| | - R Ciérvide
- Department of Radiation Oncology, HM Hospitales, Calle Oña 10, 28050, Madrid, Spain
| | - E Sánchez
- Department of Radiation Oncology, HM Hospitales, Calle Oña 10, 28050, Madrid, Spain
| | - O Hernando
- Department of Radiation Oncology, HM Hospitales, Calle Oña 10, 28050, Madrid, Spain
| | - M García-Aranda
- Department of Radiation Oncology, HM Hospitales, Calle Oña 10, 28050, Madrid, Spain
| | - J Martí
- Department of Medical Physics, HM Hospitales, Madrid, Spain
| | - A Prado
- Department of Medical Physics, HM Hospitales, Madrid, Spain
| | - X Chen-Zhao
- Department of Radiation Oncology, HM Hospitales, Calle Oña 10, 28050, Madrid, Spain
| | - C Rubio
- Department of Radiation Oncology, HM Hospitales, Calle Oña 10, 28050, Madrid, Spain
| |
Collapse
|
16
|
Hasan HF, Mostafa DM, Lotfy DM. Concerted hepatoprotective effect of bradykinin potentiating factor and low dose of γ- radiation on Naja haje envenomed rats via Bax/Bcl2 pathway. Toxicol Mech Methods 2021; 32:67-76. [PMID: 34353221 DOI: 10.1080/15376516.2021.1965276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
This study investigates the concerted hepatoprotective effects for three doses of bradykinin potentiating factor (BPF) and/or followed by exposure to a low dose of γ-radiation (LDR) against Naja haje envenoming in rats. Male rats were injected with three consecutive doses of BPF (1 μg/g i.p. for 3 days), followed by exposure to a low dose of gamma radiation (0.5 Gy), and then rats were injected with a dose of Naja haje venom (250 μg/kg i.p.). Results showed that Naja haje causes liver damage, significant elevation of alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), cytochrome c, Nitric oxide (NO), malondialdehyde (MDA) and significant depletion in glutathione peroxidase (GPx) contents. In addition, significant depletion in B-cell lymphoma 2 (Bcl-2) and significant elevation in BcL-2 associated X (Bax protein), nuclear factor kappa B (NF-κB), interleukin-1β (IL-1β) in hepatocytes. Bradykinin potentiating factor and/or low dose of γ-radiation caused improvement in liver damage caused by Naja haje venom by a significant decrease in ALT, AST, ALP levels, Bax, cytochrome c, NF-κB, IL-1β, NO and MDA contents, BPF alone or combined with low dose radiation caused a significant increase in Bcl2 and GPx contents. In conclusion, the concerted impact of BPF and LDR may provide an effective venom detoxification tool that helps to reduce hepatic toxicity and extends the lifespan.
Collapse
Affiliation(s)
- Hesham Farouk Hasan
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Dalia M Mostafa
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Dina Mahmoud Lotfy
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| |
Collapse
|
17
|
Low-dose radiotherapy for painful osteoarthritis of the elderly: A multicenter analysis of 970 patients with 1185 treated sites. Strahlenther Onkol 2021; 197:895-902. [PMID: 34342662 PMCID: PMC8458208 DOI: 10.1007/s00066-021-01816-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/01/2021] [Indexed: 10/25/2022]
Abstract
PURPOSE Painful osteoarthritis is common in elderly patients, and low-dose radiotherapy has been demonstrated to provide effective symptomatic treatment. We examined the analgesic effects of low-dose radiotherapy for osteoarthritis in the elderly aiming to reveal potential differences in the response rates relating to increasing age. METHODS A retrospective analysis was performed at two university hospitals including elderly patients (≥ 65 years) undergoing radiotherapy for osteoarthritis between 2008 and 2020. Pain intensity and response were quantified using the numerical rating scale (NRS) and the Pannewitz score. Age groups were defined for young old (65-74 years), older old (75-84 years), and oldest old patients (≥ 85 years). RESULTS In all, 970 patients with 1185 treated sites and a median age of 76 years were analyzed. Mean NRS was 66 at baseline (t0), 53 after radiotherapy (t1), and 44 at first follow-up (t2) (p < 0.001 for t0-t1, t1-t2, and t0-t2). At t1, 1.5% exhibited a Pannewitz score of 0 (no pain), 58.5% of 1-2 (less pain), 36.1% of 3 (equal pain), and 3.9% of 4 (worse pain), while at t2, pain response shifted towards 6.9% (0), 58.6% (1-2), 28.1% (3), and 6.3% (4). Pain response did not differ between age groups at t1 (p = 0.172) or t2 (p = 0.684). In addition, pain response after re-irradiation (n = 384 sites) was 61.0% and was comparable between age groups (p = 0.535). CONCLUSION Low-dose radiotherapy results in pain reduction in about two-thirds of treated sites with no difference relating to increasing age, showing that radiotherapy is an effective analgesic treatment for osteoarthritis even at advanced ages.
Collapse
|
18
|
Meziani L, Robert C, Classe M, Da Costa B, Mondini M, Clémenson C, Alfaro A, Mordant P, Ammari S, Le Goffic R, Deutsch E. Low Doses of Radiation Increase the Immunosuppressive Profile of Lung Macrophages During Viral Infection and Pneumonia. Int J Radiat Oncol Biol Phys 2021; 110:1283-1294. [PMID: 33722770 PMCID: PMC7954779 DOI: 10.1016/j.ijrobp.2021.03.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/02/2022]
Abstract
Purpose Severe pneumonia and acute respiratory distress syndrome (ARDS) have been described in patients with severe coronavirus disease 2019 (COVID-19). Recently, early clinical data reported the feasibility of low doses of radiation therapy (RT) in the treatment of ARDS in patients with severe COVID-19. However, the involved mechanisms remained unknown. Methods and Materials Here, we used airways-instilled lipopolysaccharide (LPS) and influenza virus (H1N1) as murine models of pneumonia, and toll-like receptor (TLR)-3 stimulation in human lung macrophages. Results Low doses of RT (0.5-1 Gray) decreased LPS-induced pneumonia, and increased the percentage of nerve- and airway-associated macrophages producing interleukin (IL) 10. During H1N1 viral infection, we observed decreased lung tissue damage and immune cell infiltration in irradiated animals. Low doses of RT increased IL-10 production by infiltrating immune cells into the lung. Irradiation of TLR-3 ligand-stimulated human lung macrophages ex vivo increased IL-10 secretion and decreased interferon γ production in the culture supernatant. The percentage of human lung macrophages producing IL-6 was also decreased. Conclusions Our data highlight a mechanism by which low doses of RT regulate lung inflammation and skew lung macrophages toward an anti-inflammatory profile. These data provide a preclinical mechanistic support to clinical trials evaluating low doses of RT, such as COVID-19-induced ARDS.
Collapse
Affiliation(s)
- Lydia Meziani
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France; Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France.
| | - Charlotte Robert
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France; Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France; Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Marion Classe
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France; Department of Pathology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Bruno Da Costa
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Michele Mondini
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France; Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France
| | - Céline Clémenson
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France; Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France
| | - Alexia Alfaro
- Gustave Roussy, Plateforme Imagerie et Cytométrie, UMS 23/3655, Université Paris-Saclay, Villejuif, France
| | - Pierre Mordant
- Department of Vascular Surgery, Thoracic Surgery, and Lung Transplantation, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, INSERM U1152, Université de Paris, Paris, France
| | - Samy Ammari
- Department of Radiology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France; BioMaps (UMR1281), Université Paris-Saclay, CNRS, INSERM, CEA, Orsay, 91471, France
| | - Ronan Le Goffic
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Eric Deutsch
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France; Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France; Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France.
| |
Collapse
|
19
|
How Macrophages Become Transcriptionally Dysregulated: A Hidden Impact of Antitumor Therapy. Int J Mol Sci 2021; 22:ijms22052662. [PMID: 33800829 PMCID: PMC7961970 DOI: 10.3390/ijms22052662] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are the essential components of the tumor microenvironment. TAMs originate from blood monocytes and undergo pro- or anti-inflammatory polarization during their life span within the tumor. The balance between macrophage functional populations and the efficacy of their antitumor activities rely on the transcription factors such as STAT1, NF-κB, IRF, and others. These molecular tools are of primary importance, as they contribute to the tumor adaptations and resistance to radio- and chemotherapy and can become important biomarkers for theranostics. Herein, we describe the major transcriptional mechanisms specific for TAM, as well as how radio- and chemotherapy can impact gene transcription and functionality of macrophages, and what are the consequences of the TAM-tumor cooperation.
Collapse
|
20
|
Venkatesulu BP, Lester S, Hsieh CE, Verma V, Sharon E, Ahmed M, Krishnan S. Low-Dose Radiation Therapy for COVID-19: Promises and Pitfalls. JNCI Cancer Spectr 2021; 5:pkaa103. [PMID: 33437924 PMCID: PMC7717342 DOI: 10.1093/jncics/pkaa103] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/24/2022] Open
Abstract
The coronavirus disease-2019 (COVID-19) pandemic caused by SARS-CoV-2 has exacted an enormous toll on healthcare systems worldwide. The cytokine storm that follows pulmonary infection is causally linked to respiratory compromise and mortality in the majority of patients. The sparsity of viable treatment options for this viral infection and the sequelae of pulmonary complications have fueled the quest for new therapeutic considerations. One such option, the long-forgotten idea of using low-dose radiation therapy, has recently found renewed interest in many academic centers. We outline the scientific and logistical rationale for consideration of this option and the mechanistic underpinnings of any potential therapeutic value, particularly as viewed from an immunological perspective. We also discuss the preliminary and/or published results of prospective trials examining low-dose radiation therapy for COVID-19.
Collapse
Affiliation(s)
- Bhanu P Venkatesulu
- Department of Radiation Oncology, Loyola University Stritch School of Medicine, Chicago, IL, USA
| | - Scott Lester
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Cheng-En Hsieh
- Department of Immunology, MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek Verma
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Elad Sharon
- Radiation Research Program, Division Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Mansoor Ahmed
- Cancer Therapy Evaluation Program, Division Cancer Treatment and Diagnosis National Cancer Institute, Bethesda, MD, USA
| | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, FL, USA
| |
Collapse
|
21
|
Chen Y, Jin H, Song Y, Huang T, Cao J, Tang Q, Zou Z. Targeting tumor-associated macrophages: A potential treatment for solid tumors. J Cell Physiol 2020; 236:3445-3465. [PMID: 33200401 DOI: 10.1002/jcp.30139] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAMs) in solid tumors exert protumor activities by releasing cytokines or growth factors into the tumor microenvironment. Increasing studies have also shown that TAMs play a key role in tumor progression, such as tumor angiogenesis, immunosuppression, cell proliferation, migration, invasion, and metastasis. A large body of evidence shows that the abundance of TAMs in solid tumors is correlated with poor disease prognosis and resistance to therapies. Therefore, targeting TAMs in solid tumors is considered to be a promising immunotherapeutic strategy. At present, the therapeutic strategies of targeting macrophages mainly include limiting monocyte recruitment, depletion strategies, promoting macrophage phagocytic activity, and induction of macrophage reprogramming. Additionally, targeting TAMs in combination with conventional therapies has been demonstrated to be a promising therapeutic strategy in solid tumors. In the present review, we summarized various TAMs-targeting therapeutic strategies for treating solid tumors. This review also discusses the challenges for targeting TAMs as tumor treatments, the obstacles in clinical trials, and the perspective for the future development of TAMs-targeting therapies for various cancers.
Collapse
Affiliation(s)
- Yibing Chen
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Huan Jin
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Yucen Song
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ting Huang
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Jun Cao
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Qing Tang
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Zhengzhi Zou
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| |
Collapse
|
22
|
Abdus-Salam AA, Olabumuyi AA, Jimoh MA, Folorunso SA, Orekoya AA. The role of radiation treatment in the management of inflammatory musculoskeletal conditions: a revisit. Radiat Oncol J 2020; 38:151-161. [PMID: 33012142 PMCID: PMC7533403 DOI: 10.3857/roj.2020.00178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/03/2020] [Indexed: 01/28/2023] Open
Abstract
Inflammatory musculoskeletal conditions are a common group of diseases among the elderly, worldwide. They are characterized by articular degenerative changes accompanied with often debilitating pain. Treatments often involve life-long analgesic therapy or joint replacement in extreme cases. The aim of this current review is to look at the role of radiation treatment with the hope of further study into the effectiveness of radiation treatment in reducing pain, eliminate or reduce the need for life-long analgesic therapy and thereby avoiding the analgesics’ side effects. Extensive literature search was done on PubMed and other available data base and the findings are presented and discussed. Literature showed that many countries in Europe, especially Germany use radiation routinely for the treatment of many degenerative disorders including osteoarthritis with good results and few side effects. A pilot study is therefore recommended with a view to establish the effectiveness or otherwise of this treatment method in patients.
Collapse
Affiliation(s)
| | | | - Mutiu Alani Jimoh
- Department of Radiation Oncology, University of Ibadan, Ibadan, Nigeria
| | | | | |
Collapse
|
23
|
Malfitano AM, Pisanti S, Napolitano F, Di Somma S, Martinelli R, Portella G. Tumor-Associated Macrophage Status in Cancer Treatment. Cancers (Basel) 2020; 12:cancers12071987. [PMID: 32708142 PMCID: PMC7409350 DOI: 10.3390/cancers12071987] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor-associated macrophages (TAMs) represent the most abundant innate immune cells in tumors. TAMs, exhibiting anti-inflammatory phenotype, are key players in cancer progression, metastasis and resistance to therapy. A high TAM infiltration is generally associated with poor prognosis, but macrophages are highly plastic cells that can adopt either proinflammatory/antitumor or anti-inflammatory/protumor features in response to tumor microenvironment stimuli. In the context of cancer therapy, many anticancer therapeutics, apart from their direct effect on tumor cells, display different effects on TAM activation status and density. In this review, we aim to evaluate the indirect effects of anticancer therapies in the modulation of TAM phenotypes and pro/antitumor activity.
Collapse
Affiliation(s)
- Anna Maria Malfitano
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.N.); (S.D.S.)
- Correspondence: (A.M.M.); (G.P.); Tel.: +39-081-746-3056 (G.P.)
| | - Simona Pisanti
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvador Allende, Baronissi, 84081 Salerno, Italy; (S.P.); (R.M.)
| | - Fabiana Napolitano
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.N.); (S.D.S.)
| | - Sarah Di Somma
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.N.); (S.D.S.)
| | - Rosanna Martinelli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvador Allende, Baronissi, 84081 Salerno, Italy; (S.P.); (R.M.)
| | - Giuseppe Portella
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.N.); (S.D.S.)
- Correspondence: (A.M.M.); (G.P.); Tel.: +39-081-746-3056 (G.P.)
| |
Collapse
|
24
|
Algara M, Arenas M, Marin J, Vallverdu I, Fernandez-Letón P, Villar J, Fabrer G, Rubio C, Montero A. Low dose anti-inflammatory radiotherapy for the treatment of pneumonia by covid-19: A proposal for a multi-centric prospective trial. Clin Transl Radiat Oncol 2020; 24:29-33. [PMID: 32613089 PMCID: PMC7317159 DOI: 10.1016/j.ctro.2020.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/06/2020] [Accepted: 06/12/2020] [Indexed: 01/20/2023] Open
Abstract
Interstitial bilateral pneumonia is the main complication of severe COVID-19. Low-dose radiation therapy (LD-RT) has well-known anti-inflammatory effects. LD-RT can interfere with the hyper-inflammatory cascade associated with severe COVID-19. LD-RT could reduce the severity of associated cytokine release caused by COVID-19. This trial will evaluate efficacy of lung LD-RT for interstitial COVID-19 pneumonia.
Background COVID-19 is a highly contagious viral infection with high morbidity that is draining health resources. The biggest complication is pneumonia, which has a serious inflammatory component, with no standardized treatment. Low-dose radiation therapy (LD-RT) is non-invasive and has anti-inflammatory effects that can interfere with the inflammatory cascade, thus reducing the severity of associated cytokine release and might be useful in the treatment of respiratory complications caused by COVID-19. Study design and methods This multicentric prospective clinical trial seeks to evaluate the efficacy of bilateral lung LD-RT therapy as a treatment for interstitial pneumonia in patients with COVID-19 for improving respiratory function. This prospective study will have 2 phases: I) an exploratory phase enrolling 10 patients, which will assess the feasibility and efficacy of low-dose lung irradiation, evaluated according to an increase in the PaO2/FiO2 ratio of at least 20% at 48–72 h with respect to the pre-irradiation value. If a minimum efficiency of 30% of the patients is not achieved, the study will not be continued. II) Non-randomized comparative phase in two groups: a control group, which will only receive pharmacological treatment, and an experimental arm with pharmacological treatment and LD-RT. It will include 96 patients, the allocation will be 1: 2, that is, 32 in the control arm and 64 in the experimental arm. The primary end-point will be the efficacy of LD-RT in patients with COVID-19 pneumonia according to an improvement in PaO2/FiO2. Secondary objectives will include the safety of bilateral lung LD-RT, an improvement in the radiology image, overall mortality rates at 15 and 30 days after irradiation and characterizing anti-inflammatory mechanisms of LD-RT by measuring the level of expression of adhesion molecules, anti-inflammatory cytokines and oxidative stress mediators. Trial registration: ClinicalTrial.gov NCT-04380818 https://clinicaltrials.gov/ct2/show/NCT04380818?term=RADIOTHERAPY&cond=COVID&draw=2&rank=4.
Collapse
Affiliation(s)
- M. Algara
- Department of Radiation Oncology, Hospital del Mar, Autonomous University of Barcelona, Spain
| | - M. Arenas
- Department of Radiation Oncology, University Hospital Sant Joan de Reus, Rovira iV irgili University, Tarragona, Spain
| | - J. Marin
- Intensive Care Unit, Hospital del Mar, Barcelona, Spain
| | - I. Vallverdu
- Intensive Care Unit University Hospital Sant Joan de Reus, Universitat Rovira I Virgili, Tarragona, Spain
| | - P. Fernandez-Letón
- Department of Radiation Physics, University Hospital HM Sanchinarro, Madrid, Spain
| | - J. Villar
- Department of Infectious Diseases, Hospital del Mar, Barcelona, Spain
| | - G. Fabrer
- Department of Geriatric and Palliative Care, University Hospital Sant Joan de Reus, Rovira i Virgili University, Tarragona, Spain
| | - C. Rubio
- Department of Radiation Oncology, University Hospital HM Sanchinarro, Madrid, Spain
| | - A. Montero
- Department of Radiation Oncology, University Hospital HM Sanchinarro, Madrid, Spain
- Corresponding author.
| |
Collapse
|
25
|
Montero A, Arenas M, Algara M. Low-dose radiation therapy: could it be a game-changer for COVID-19? Clin Transl Oncol 2020; 23:1-4. [PMID: 32451973 PMCID: PMC7247741 DOI: 10.1007/s12094-020-02401-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 05/12/2020] [Indexed: 12/02/2022]
Affiliation(s)
- A Montero
- Oncología Radioterápica, Hospital Universitario HM Sanchinarro, Madrid, Spain.
| | - M Arenas
- Oncología Radioterápica, Hospital Universitario Sant Joan de Reus, Universitat Rovira I Virgili, Reus, Spain
| | - M Algara
- Oncología Radioterápica, Hospital de la Esperanza, Parc de Salut del Mar, Universitat Autónoma de Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Pandey VK, Shankar BS. Radiation-induced augmentation in dendritic cell function is mediated by apoptotic bodies/STAT5/Zbtb46 signaling. Int J Radiat Biol 2020; 96:988-998. [PMID: 32396024 DOI: 10.1080/09553002.2020.1767818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Purpose: To evaluate the effect of ionizing radiation (IR) exposure on differentiation and maturation of dendritic cells (DC).Materials and methods: Bone marrow progenitor cells irradiated in vitro or isolated from mice exposed to whole body or localized tumor irradiation were differentiated into DC. Phenotypic maturation of DC was characterized by labeling with specific antibodies and flow cytometry analysis. Cytokines were estimated by ELISA.Results: Splenic and bone marrow-derived DC (BMDC) from tumor-bearing mice exposed to localized irradiation showed abrogation of tumor-induced immunosuppression. This was not due to the effect of radiation on tumor cells as DC derived from normal mice exposed to whole-body irradiation (WBI) also showed increase in immune-activating potential of DC. This was observed in terms of increased phenotypic and functional activation of DCs. This phenomenon was also recapitulated if DC were differentiated from in vitro irradiated progenitor cells and was found to be due to STAT5/Zbtb46 signaling mediated by the irradiation-induced apoptotic bodies (ABs). When these ABs were depleted using annexin-beads, these effects were reversed confirming the involvement of this pathway. The role of ABs was further validated in DC derived from mice exposed to WBI in adaptive response experiments with 0.1 Gy priming dose prior to 2 Gy challenge dose. A corresponding reduction in DC maturation markers was observed with decrease in apoptosis in vivo. Further, these DCs derived from irradiated progenitors (IP) could resist the suppressive effects of tumor conditioned medium (TCM) and had increased immune-activating potential as seen in the tumor-bearing mice.Conclusions: Though radiation is the most commonly used therapeutic modality for cancer, its effects on dendritic cell differentiation is not completely understood. We demonstrate here for the first time that exposure to select doses of IR can increase immune-activating potential of DC through ABs. This can have implications in selection of appropriate doses of IR during radiotherapy of cancer patients.
Collapse
Affiliation(s)
- Vipul K Pandey
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Bhavani S Shankar
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
27
|
El-Ghazaly MA, Fadel NA, Abdel-Naby DH, Abd El-Rehim HA, Zaki HF, Kenawy SA. Amelioration of adjuvant-induced arthritis by exposure to low dose gamma radiation and resveratrol administration in rats. Int J Radiat Biol 2020; 96:857-867. [PMID: 32216648 DOI: 10.1080/09553002.2020.1748911] [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] [Indexed: 12/13/2022]
Abstract
Purpose: Low dose radiation has been reported as an effective treatment for rheumatoid arthritis via multiple dose exposures. The present study was designed to increase the therapeutic efficacy of low dose radiation with the minimum exposure level in arthritic rats by concurrent administration of resveratrol (RSV) as an adjunctive therapy with anti-inflammatory properties.Materials and methods: Rats were rendered arthritic by sub-plantar injection of Freund's complete adjuvant (FCA) and exposed to low dose radiation at a total exposure level of 0.5 Gy (2 × 0.25). During the exposure course, RSV (50 mg/kg) was orally administered once daily for two weeks. Diclofenac (3 mg/kg) was administered as a standard anti-inflammatory drug. Paw volume was measured every 4 days. After 28 days of induction, rats were sacrificed and serum was collected for estimation of tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1β), thiobarbituric acid reactive substances (TBARS), and total nitrate/nitrite (NOx). Furthermore, paws were dissected for histopathological examinations and immuno-histochemical estimation of nuclear factor-kappa B p65 (NF-κB p65) expression.Results: Administration of RSV during the low dose radiation exposure course produced a significant decrease in the paw swelling and a potentiated inhibition in the serum levels of TNF-α, IL-1β, TBARs, and NOx. The dual treatment strategy alleviated the histopathological damage to a greater extent than that produced by each treatment. Moreover, a pronounced suppression of NF-κB p65 expression in the synovial tissue was observed in the combination group. The combination treatment showed a nearly similar potency to that observed in the diclofenac treated group.Conclusion: Administration of RSV augmented the modulatory activity of low dose radiation with minimum exposure level on the disease progression.
Collapse
Affiliation(s)
- Mona A El-Ghazaly
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Noha A Fadel
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Doaa H Abdel-Naby
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Hassan A Abd El-Rehim
- Department of Polymers, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Sanaa A Kenawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
28
|
Álvarez B, Montero Á, Aramburu F, Calvo E, Ángel de la Casa M, Valero J, Hernando O, López M, Ciérvide R, García-Aranda M, Rodríguez S, Sánchez E, Chen X, Alonso R, García de la Peña P, Rubio C. Radiotherapy for ostheoarticular degenerative disorders: When nothing else works. OSTEOARTHRITIS AND CARTILAGE OPEN 2020; 1:100016. [DOI: 10.1016/j.ocarto.2019.100016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 11/27/2019] [Indexed: 01/05/2023] Open
|
29
|
Radiobiological Principles of Radiotherapy for Benign Diseases. Radiat Oncol 2020. [DOI: 10.1007/978-3-319-52619-5_133-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
30
|
Low-Dose Irradiation Differentially Impacts Macrophage Phenotype in Dependence of Fibroblast-Like Synoviocytes and Radiation Dose. J Immunol Res 2019; 2019:3161750. [PMID: 31485459 PMCID: PMC6710796 DOI: 10.1155/2019/3161750] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/01/2019] [Accepted: 07/11/2019] [Indexed: 12/17/2022] Open
Abstract
Rheumatoid arthritis (RA) is a multifactorial autoimmune disease whose main hallmark is inflammation and destruction of the joints. Two cell types within the synovium that play an important role in RA are fibroblast-like synoviocytes (FLS) and macrophages. The latter innate immune cells show a high plasticity in their phenotype and are central in inflammatory processes. Low-dose radiotherapy (LD-RT) with particularly a single dose of 0.5 Gy has been demonstrated to have a positive impact on pain, inflammation, and bone in inflamed joints. We now examined for the first time how LD-RT influences FLS and bone marrow-derived macrophages in co-culture systems of an experimental model of RA to reveal further mechanisms of immune modulatory effects of low and intermediate dose of ionizing radiation. For this, the bone marrow of hTNF-α tg mice was differentiated either with cytokines to obtain key macrophage phenotypes (M0, M1, and M2) or with supernatants (SN) of untreated or irradiated FLS. Flow cytometry analyses were used to analyse the impact of radiation (0.1, 0.5, 1.0, and 2.0 Gy) on the phenotype of macrophages in the presence or absence of SN of FLS. LD-RT had no impact on cytokine-mediated macrophage polarization in M0, M1, or M2 macrophages. However, SN of irradiated FLS particularly reduced CD206 expression on macrophages. Macrophage phenotype was stable when being in contact with SN of nonirradiated FLS, but significantly increased surface expression of CD206 and slightly decreased CD80 and CD86 expression were observed when macrophage themselves were irradiated with 0.5 Gy under these microenvironmental conditions, again highlighting discontinuous dose dependencies in the low and intermediate dose range. One can conclude that FLS-dependent microenvironmental conditions have a slight influence on the modulation of macrophage phenotype under radiation exposure conditions. Future studies are needed to reveal the impact of radiation exposure on the functions of treated macrophages under such microenvironmental conditions.
Collapse
|
31
|
Guéguen Y, Bontemps A, Ebrahimian TG. Adaptive responses to low doses of radiation or chemicals: their cellular and molecular mechanisms. Cell Mol Life Sci 2019; 76:1255-1273. [PMID: 30535789 PMCID: PMC11105647 DOI: 10.1007/s00018-018-2987-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/09/2018] [Accepted: 12/03/2018] [Indexed: 12/17/2022]
Abstract
This article reviews the current knowledge on the mechanisms of adaptive response to low doses of ionizing radiation or chemical exposure. A better knowledge of these mechanisms is needed to improve our understanding of health risks at low levels of environmental or occupational exposure and their involvement in cancer or non-cancer diseases. This response is orchestrated through a multifaceted cellular program involving the concerted action of diverse stress response pathways. These evolutionary highly conserved defense mechanisms determine the cellular response to chemical and physical aggression. They include DNA damage repair (p53, ATM, PARP pathways), antioxidant response (Nrf2 pathway), immune/inflammatory response (NF-κB pathway), cell survival/death pathway (apoptosis), endoplasmic response to stress (UPR response), and other cytoprotective processes including autophagy, cell cycle regulation, and the unfolded protein response. The coordinated action of these processes induced by low-dose radiation or chemicals produces biological effects that are currently estimated with the linear non-threshold model. These effects are controversial. They are difficult to detect because of their low magnitude, the scarcity of events in humans, and the difficulty of corroborating associations over the long term. Improving our understanding of these biological consequences should help humans and their environment by enabling better risk estimates, the revision of radiation protection standards, and possible therapeutic advances.
Collapse
Affiliation(s)
- Yann Guéguen
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE, SESANE, LRTOX, B.P. no 17, 92262, Fontenay-aux-Roses Cedex, France.
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE, SESANE, LRSI, Fontenay-aux-Roses, France.
| | - Alice Bontemps
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE, SESANE, LRTOX, B.P. no 17, 92262, Fontenay-aux-Roses Cedex, France
| | - Teni G Ebrahimian
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE, SESANE, LRTOX, B.P. no 17, 92262, Fontenay-aux-Roses Cedex, France
| |
Collapse
|
32
|
Calabrese EJ, Giordano JJ, Kozumbo WJ, Leak RK, Bhatia TN. Hormesis mediates dose-sensitive shifts in macrophage activation patterns. Pharmacol Res 2018; 137:236-249. [DOI: 10.1016/j.phrs.2018.10.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 02/07/2023]
|
33
|
Wunderlich R, Rühle PF, Deloch L, Rödel F, Fietkau R, Gaipl US, Frey B. Ionizing radiation reduces the capacity of activated macrophages to induce T-cell proliferation, but does not trigger dendritic cell-mediated non-targeted effects. Int J Radiat Biol 2018; 95:33-43. [DOI: 10.1080/09553002.2018.1490037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Roland Wunderlich
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, Neuherberg, Germany
| | - Paul Friedrich Rühle
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lisa Deloch
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt am Main, Frankfurt, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Udo S. Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
34
|
Martinez-Zubiaurre I, Chalmers AJ, Hellevik T. Radiation-Induced Transformation of Immunoregulatory Networks in the Tumor Stroma. Front Immunol 2018; 9:1679. [PMID: 30105016 PMCID: PMC6077256 DOI: 10.3389/fimmu.2018.01679] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/09/2018] [Indexed: 12/27/2022] Open
Abstract
The implementation of novel cancer immunotherapies in the form of immune checkpoint blockers represents a major advancement in the treatment of cancer, and has renewed enthusiasm for identifying new ways to induce antitumor immune responses in patients. Despite the proven efficacy of neutralizing antibodies that target immune checkpoints in some refractory cancers, many patients do not experience therapeutic benefit, possibly owing to a lack of antitumor immune recognition, or to the presence of dominant immunosuppressive mechanisms in the tumor microenvironment (TME). Recent developments in this field have revealed that local radiotherapy (RT) can transform tumors into in situ vaccines, and may help to overcome some of the barriers to tumor-specific immune rejection. RT has the potential to ignite tumor immune recognition by generating immunogenic signals and releasing neoantigens, but the multiple immunosuppressive forces in the TME continue to represent important barriers to successful tumor rejection. In this article, we review the radiation-induced changes in the stromal compartments of tumors that could have an impact on tumor immune attack. Since different RT regimens are known to mediate strikingly different effects on the multifarious elements of the tumor stroma, special emphasis is given to different RT schedules, and the time after treatment at which the effects are measured. A better understanding of TME remodeling following specific RT regimens and the window of opportunity offered by RT will enable optimization of the design of novel treatment combinations.
Collapse
Affiliation(s)
- Inigo Martinez-Zubiaurre
- Department of Clinical Medicine, Faculty of Health Sciences, UiT the Arctic University of Norway, Tromsø, Norway
| | - Anthony J Chalmers
- Institute of Cancer Sciences, Beatson West of Scotland Cancer Centre, University of Glasgow, Glasgow, United Kingdom
| | - Turid Hellevik
- Department of Radiation Oncology, University Hospital of Northern Norway, Tromsø, Norway
| |
Collapse
|
35
|
Frey B, Rückert M, Deloch L, Rühle PF, Derer A, Fietkau R, Gaipl US. Immunomodulation by ionizing radiation-impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunol Rev 2018; 280:231-248. [PMID: 29027224 DOI: 10.1111/imr.12572] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ionizing radiation is often regarded as an element of danger. But, danger responses on the cellular and molecular level are often beneficial with regard to the induction of anti-tumor immunity and for amelioration of inflammation. We outline how in dependence of radiation dose and fraction, radiation itself-and especially in combination with immune modulators-impacts on the innate and adaptive immune system. Focus is set on radiation-induced changes of the tumor cell phenotype and the cellular microenvironment including immunogenic cancer cell death. Mechanisms how anti-tumor immune responses are triggered by radiotherapy in combination with hyperthermia, inhibition of apoptosis, the adjuvant AnnexinA5, or vaccination with high hydrostatic pressure-killed autologous tumor cells are discussed. Building on this, feasible multimodal radio-immunotherapy concepts are reviewed including overcoming immune suppression by immune checkpoint inhibitors and by targeting TGF-β. Since radiation-induced tissue damage, inflammation, and anti-tumor immune responses are interconnected, the impact of lower doses of radiation on amelioration of inflammation is outlined. Closely meshed immune monitoring concepts based on the liquid biopsy blood are suggested for prognosis and prediction of cancer and non-cancer inflammatory diseases. Finally, challenges and visions for the design of cancer radio-immunotherapies and for treatment of benign inflammatory diseases are given.
Collapse
Affiliation(s)
- Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Michael Rückert
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lisa Deloch
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Paul F Rühle
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anja Derer
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
36
|
Averbeck D, Salomaa S, Bouffler S, Ottolenghi A, Smyth V, Sabatier L. Progress in low dose health risk research. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 776:46-69. [DOI: 10.1016/j.mrrev.2018.04.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
|
37
|
Rückert M, Deloch L, Fietkau R, Frey B, Hecht M, Gaipl US. Immune modulatory effects of radiotherapy as basis for well-reasoned radioimmunotherapies. Strahlenther Onkol 2018; 194:509-519. [PMID: 29500551 DOI: 10.1007/s00066-018-1287-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/19/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Radiotherapy (RT) has been known for decades as a local treatment modality for malign and benign disease. In order to efficiently exploit the therapeutic potential of RT, an understanding of the immune modulatory properties of ionizing radiation is mandatory. These should be used for improvement of radioimmunotherapies for cancer in particular. METHODS We here summarize the latest research and review articles about immune modulatory properties of RT, with focus on radiation dose and on combination of RT with selected immunotherapies. Based on the knowledge of the manifold immune mechanisms that are triggered by RT, thought-provoking impulse for multimodal radioimmunotherapies is provided. RESULTS It has become obvious that ionizing radiation induces various forms of cell death and associated processes via DNA damage initiation and triggering of cellular stress responses. Immunogenic cell death (ICD) is of special interest since it activates the immune system via release of danger signals and via direct activation of immune cells. While RT with higher single doses in particular induces ICD, RT with a lower dose is mainly responsible for immune cell recruitment and for attenuation of an existing inflammation. The counteracting immunosuppression emanating from tumor cells can be overcome by combining RT with selected immunotherapies such as immune checkpoint inhibition, TGF-β inhibitors, and boosting of immunity with vaccination. CONCLUSION In order to exploit the full power of RT and thereby develop efficient radioimmunotherapies, the dose per fraction used in RT protocols, the fractionation, the quality, and the quantity of certain immunotherapies need to be qualitatively and chronologically well-matched to the individual immune status of the patient.
Collapse
Affiliation(s)
- Michael Rückert
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstr. 27, 91054, Erlangen, Germany
| | - Lisa Deloch
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstr. 27, 91054, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstr. 27, 91054, Erlangen, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstr. 27, 91054, Erlangen, Germany
| | - Markus Hecht
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstr. 27, 91054, Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstr. 27, 91054, Erlangen, Germany.
| |
Collapse
|
38
|
Ebrahimian TG, Beugnies L, Surette J, Priest N, Gueguen Y, Gloaguen C, Benderitter M, Jourdain JR, Tack K. Chronic Exposure to External Low-Dose Gamma Radiation Induces an Increase in Anti-inflammatory and Anti-oxidative Parameters Resulting in Atherosclerotic Plaque Size Reduction in ApoE -/- Mice. Radiat Res 2017; 189:187-196. [PMID: 29227739 DOI: 10.1667/rr14823.1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Populations living in radiation-contaminated territories, such as Chernobyl and Fukushima, are chronically exposed to external gamma radiation and internal radionuclide contamination due to the large amount of 137Cs released in the environment. The effect of chronic low-dose exposure on the development of cardiovascular diseases remains unclear. Previously reported studies have shown that low-dose radiation exposure could lead to discrepancies according to dose rate. In this study, we examined the effect of very low-dose and dose-rate chronic external exposure on atherosclerosis development. ApoE-/- mice were chronically irradiated with a gamma source for 8 months at two different dose rates, 12 and 28 μGy/h, equivalent to dose rates measured in contaminated territories, with a cumulative dose of 67 and 157 mGy, respectively. We evaluated plaque size and phenotype, inflammatory profile and oxidative stress status. The results of this study showed a decrease in plaque sizes and an increase in collagen content in ApoE-/- mice exposed to 28 μGy/h for 8 months compared to nonexposed animals. The plaque phenotype was associated with an increase in anti-inflammatory and anti-oxidative gene expression. These results suggest that chronic low-dose gamma irradiation induces an upregulation of organism defenses leading to a decrease in inflammation and plaque size. To our knowledge, this is the first study to describe the possible effect of chronic external very low-dose ionizing radiation exposure for 8 months. This work could help to identify the potential existence of a dose threshold, below that which harmful effects are not exhibited and beneficial effects are potentially observed. Furthermore, these findings permit consideration of the importance of dose rate in radiation protection.
Collapse
Affiliation(s)
- T G Ebrahimian
- a Institut de Radioprotection et de Sureté Nucléaire (IRSN), PRP-HOM, SRBE, Fontenay-aux-Roses, France; and
| | - L Beugnies
- a Institut de Radioprotection et de Sureté Nucléaire (IRSN), PRP-HOM, SRBE, Fontenay-aux-Roses, France; and
| | - J Surette
- b Radiobiology and Health, Canadian Nuclear Laboratories, Chalk-River, Ontario K0J 1J0, Canada
| | - N Priest
- b Radiobiology and Health, Canadian Nuclear Laboratories, Chalk-River, Ontario K0J 1J0, Canada
| | - Y Gueguen
- a Institut de Radioprotection et de Sureté Nucléaire (IRSN), PRP-HOM, SRBE, Fontenay-aux-Roses, France; and
| | - C Gloaguen
- a Institut de Radioprotection et de Sureté Nucléaire (IRSN), PRP-HOM, SRBE, Fontenay-aux-Roses, France; and
| | - M Benderitter
- a Institut de Radioprotection et de Sureté Nucléaire (IRSN), PRP-HOM, SRBE, Fontenay-aux-Roses, France; and
| | - J R Jourdain
- a Institut de Radioprotection et de Sureté Nucléaire (IRSN), PRP-HOM, SRBE, Fontenay-aux-Roses, France; and
| | - K Tack
- a Institut de Radioprotection et de Sureté Nucléaire (IRSN), PRP-HOM, SRBE, Fontenay-aux-Roses, France; and
| |
Collapse
|
39
|
Li J, Yao ZY, She C, Li J, Ten B, Liu C, Lin SB, Dong QR, Ren PG. Effects of low-dose X-ray irradiation on activated macrophages and their possible signal pathways. PLoS One 2017; 12:e0185854. [PMID: 29077718 PMCID: PMC5659615 DOI: 10.1371/journal.pone.0185854] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 09/20/2017] [Indexed: 11/19/2022] Open
Abstract
Low-dose irradiation (LDI) has been used in clinics to treat human diseases, including chronic inflammation. This study assessed the effects of LDI on the inflammatory response of activated mouse primary peritoneal macrophages, and the underlying signal pathways. Primary peritoneal macrophages were isolated from mice and then incubated with lipopolysaccharide (LPS)-coated Ti microparticles (Ti-positive control) with or without brief exposure to LDI (X-ray, 0.5 Gy) 1 h later (Ti-LDI group) or left untreated in culture medium (Ti-negative control). The macrophages were then subjected to qRT-PCR, Western blot, cell viability CCK-8 assay, and ELISA. qRT-PCR analysis revealed the Ti-LDI group expressed significantly lower levels of IL-1β, IL-6, and TNF-α mRNA than those of the Ti-positive control group, while the ELISA data showed that Ti-LDI group had significantly lower secretion of IL-1β, IL-6, and TNF-α proteins. The most significant reduction associated with LDI was the secretion TNF-α protein, which barely increased from 13 to 25 h after treatment. Western blot data demonstrated that phosphorylation of p65 and ERK was much lower in the Ti-LDI group than in the controls. The data from the current study suggests that LDI of activated mouse macrophages was associated with significantly lower inflammation responses, compared with non-exposed activated macrophages, which was possibly through inhibition of the NF-κB and ERK pathways.
Collapse
Affiliation(s)
- Jian Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhen-yu Yao
- Department of Translational Medicine R&D Center, Shenzhen Institute of Advanced Technology, CAS, Shenzhen, Guangdong, China
| | - Chang She
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jian Li
- Department of Translational Medicine R&D Center, Shenzhen Institute of Advanced Technology, CAS, Shenzhen, Guangdong, China
| | - Bin Ten
- Department of Translational Medicine R&D Center, Shenzhen Institute of Advanced Technology, CAS, Shenzhen, Guangdong, China
| | - Chang Liu
- Department of Translational Medicine R&D Center, Shenzhen Institute of Advanced Technology, CAS, Shenzhen, Guangdong, China
| | - Shu-bin Lin
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi-Rong Dong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- * E-mail: (QRD); (PGR)
| | - Pei-Gen Ren
- Department of Translational Medicine R&D Center, Shenzhen Institute of Advanced Technology, CAS, Shenzhen, Guangdong, China
- * E-mail: (QRD); (PGR)
| |
Collapse
|
40
|
Wu Q, Allouch A, Martins I, Modjtahedi N, Deutsch E, Perfettini JL. Macrophage biology plays a central role during ionizing radiation-elicited tumor response. Biomed J 2017; 40:200-211. [PMID: 28918908 PMCID: PMC6136289 DOI: 10.1016/j.bj.2017.06.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 06/01/2017] [Accepted: 06/11/2017] [Indexed: 12/13/2022] Open
Abstract
Radiation therapy is one of the major therapeutic modalities for most solid tumors. The anti-tumor effect of radiation therapy consists of the direct tumor cell killing, as well as the modulation of tumor microenvironment and the activation of immune response against tumors. Radiation therapy has been shown to promote immunogenic cells death, activate dendritic cells and enhance tumor antigen presentation and anti-tumor T cell activation. Radiation therapy also programs innate immune cells such as macrophages that leads to either radiosensitization or radioresistance, according to different tumors and different radiation regimen studied. The mechanisms underlying radiation-induced macrophage activation remain largely elusive. Various molecular players such as NF-κB, MAPKs, p53, reactive oxygen species, inflammasomes have been involved in these processes. The skewing to a pro-inflammatory phenotype thus results in the activation of anti-tumor immune response and enhanced radiotherapy effect. Therefore, a comprehensive understanding of the mechanism of radiation-induced macrophage activation and its role in tumor response to radiation therapy is crucial for the development of new therapeutic strategies to enhance radiation therapy efficacy.
Collapse
Affiliation(s)
- Qiuji Wu
- Cell Death and Aging Team, Gystave Roussy Cancer Campus, Villejuif, France; Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France; Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Hubei, China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Hubei, China
| | - Awatef Allouch
- Cell Death and Aging Team, Gystave Roussy Cancer Campus, Villejuif, France; Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France
| | - Isabelle Martins
- Cell Death and Aging Team, Gystave Roussy Cancer Campus, Villejuif, France; Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France
| | - Nazanine Modjtahedi
- Cell Death and Aging Team, Gystave Roussy Cancer Campus, Villejuif, France; Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France
| | - Eric Deutsch
- Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France
| | - Jean-Luc Perfettini
- Cell Death and Aging Team, Gystave Roussy Cancer Campus, Villejuif, France; Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France.
| |
Collapse
|
41
|
Genard G, Lucas S, Michiels C. Reprogramming of Tumor-Associated Macrophages with Anticancer Therapies: Radiotherapy versus Chemo- and Immunotherapies. Front Immunol 2017; 8:828. [PMID: 28769933 PMCID: PMC5509958 DOI: 10.3389/fimmu.2017.00828] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/30/2017] [Indexed: 12/15/2022] Open
Abstract
Tumor-associated macrophages (TAMs) play a central role in tumor progression, metastasis, and recurrence after treatment. Macrophage plasticity and diversity allow their classification along a M1–M2 polarization axis. Tumor-associated macrophages usually display a M2-like phenotype, associated with pro-tumoral features whereas M1 macrophages exert antitumor functions. Targeting the reprogramming of TAMs toward M1-like macrophages would thus be an efficient way to promote tumor regression. This can be achieved through therapies including chemotherapy, immunotherapy, and radiotherapy (RT). In this review, we first describe how chemo- and immunotherapies can target TAMs and, second, we detail how RT modifies macrophage phenotype and present the molecular pathways that may be involved. The identification of irradiation dose inducing macrophage reprogramming and of the underlying mechanisms could lead to the design of novel therapeutic strategies and improve synergy in combined treatments.
Collapse
Affiliation(s)
- Géraldine Genard
- URBC - NARILIS, University of Namur, Namur, Belgium.,Laboratory of Analysis by Nuclear Reaction (LARN/PMR) - NARILIS, University of Namur, Namur, Belgium
| | - Stéphane Lucas
- Laboratory of Analysis by Nuclear Reaction (LARN/PMR) - NARILIS, University of Namur, Namur, Belgium
| | | |
Collapse
|
42
|
Wu Q, Allouch A, Martins I, Brenner C, Modjtahedi N, Deutsch E, Perfettini JL. Modulating Both Tumor Cell Death and Innate Immunity Is Essential for Improving Radiation Therapy Effectiveness. Front Immunol 2017; 8:613. [PMID: 28603525 PMCID: PMC5445662 DOI: 10.3389/fimmu.2017.00613] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 05/09/2017] [Indexed: 12/17/2022] Open
Abstract
Radiation therapy is one of the cornerstones of cancer treatment. In tumor cells, exposure to ionizing radiation (IR) provokes DNA damages that trigger various forms of cell death such as apoptosis, necrosis, autophagic cell death, and mitotic catastrophe. IR can also induce cellular senescence that could serve as an additional antitumor barrier in a context-dependent manner. Moreover, accumulating evidence has demonstrated that IR interacts profoundly with tumor-infiltrating immune cells, which cooperatively drive treatment outcomes. Recent preclinical and clinical successes due to the combination of radiation therapy and immune checkpoint blockade have underscored the need for a better understanding of the interplay between radiation therapy and the immune system. In this review, we will present an overview of cell death modalities induced by IR, summarize the immunogenic properties of irradiated cancer cells, and discuss the biological consequences of IR on innate immune cell functions, with a particular attention on dendritic cells, macrophages, and NK cells. Finally, we will discuss their potential applications in cancer treatment.
Collapse
Affiliation(s)
- Qiuji Wu
- Cell Death and Aging Team, Gustave Roussy Cancer Campus, Villejuif, France.,Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France.,Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Awatef Allouch
- Cell Death and Aging Team, Gustave Roussy Cancer Campus, Villejuif, France.,Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France
| | - Isabelle Martins
- Cell Death and Aging Team, Gustave Roussy Cancer Campus, Villejuif, France.,Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France
| | - Catherine Brenner
- Laboratory of Signaling and Cardiovascular Pathophysiology, INSERM UMR-S 1180, Université Paris-Sud, Faculté de Pharmacie, Châtenay-Malabry, France
| | - Nazanine Modjtahedi
- Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France
| | - Eric Deutsch
- Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France
| | - Jean-Luc Perfettini
- Cell Death and Aging Team, Gustave Roussy Cancer Campus, Villejuif, France.,Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France
| |
Collapse
|
43
|
Rödel F, Fournier C, Wiedemann J, Merz F, Gaipl US, Frey B, Keilholz L, Seegenschmiedt MH, Rödel C, Hehlgans S. Basics of Radiation Biology When Treating Hyperproliferative Benign Diseases. Front Immunol 2017; 8:519. [PMID: 28515727 PMCID: PMC5413517 DOI: 10.3389/fimmu.2017.00519] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/18/2017] [Indexed: 01/08/2023] Open
Abstract
For decades, low- and moderate-dose radiation therapy (RT) has been shown to exert a beneficial therapeutic effect in a multitude of non-malignant conditions including painful degenerative muscoloskeletal and hyperproliferative disorders. Dupuytren and Ledderhose diseases are benign fibroproliferative diseases of the hand/foot with fibrotic nodules and fascial cords, which determine debilitating contractures and deformities of fingers/toes, while keloids are exuberant scar formations following burn damage, surgery, and trauma. Although RT has become an established and effective option in the management of these diseases, experimental studies to illustrate cellular composites and factors involved remain to be elucidated. More recent findings, however, indicate the involvement of radiation-sensitive targets like mitotic fibroblasts/myofibroblasts as well as inflammatory cells. Radiation-related molecular mechanisms affecting these target cells include the production of free radicals to hamper proliferative activity and interference with growth factors and cytokines. Moreover, an impairment of activated immune cells involved in both myofibroblast proliferative and inflammatory processes may further contribute to the clinical effects. We here aim at briefly describing mechanisms contributing to a modulation of proliferative and inflammatory processes and to summarize current concepts of treating hyperproliferative diseases by low and moderate doses of ionizing radiation.
Collapse
Affiliation(s)
- Franz Rödel
- Department of Radiotherapy and Oncology, University Hospital of Frankfurt, Goethe-Universität, Frankfurt am Main, Germany
| | - Claudia Fournier
- Department of Biophysics, GSI Helmholtz Centre for Heavy Ion Research, Darmstadt, Germany
| | - Julia Wiedemann
- Department of Biophysics, GSI Helmholtz Centre for Heavy Ion Research, Darmstadt, Germany
| | - Felicitas Merz
- Department of Biophysics, GSI Helmholtz Centre for Heavy Ion Research, Darmstadt, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ludwig Keilholz
- Department of Radiotherapy, Clinical Center Bayreuth, Bayreuth, Germany
| | | | - Claus Rödel
- Department of Radiotherapy and Oncology, University Hospital of Frankfurt, Goethe-Universität, Frankfurt am Main, Germany
| | - Stephanie Hehlgans
- Department of Radiotherapy and Oncology, University Hospital of Frankfurt, Goethe-Universität, Frankfurt am Main, Germany
| |
Collapse
|
44
|
The Effect of Tacrolimus and Mycophenolic Acid on CD14+ Monocyte Activation and Function. PLoS One 2017; 12:e0170806. [PMID: 28122021 PMCID: PMC5266297 DOI: 10.1371/journal.pone.0170806] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/11/2017] [Indexed: 12/20/2022] Open
Abstract
Monocytes and macrophages play key roles in many disease states, including cellular and humoral rejection after solid organ transplantation (SOT). To suppress alloimmunity after SOT, immunosuppressive drug therapy is necessary. However, little is known about the effects of the immunosuppressive drugs tacrolimus and mycophenolic acid (MPA) on monocyte activation and function. Here, the effect of these immunosuppressants on monocytes was investigated by measuring phosphorylation of three intracellular signaling proteins which all have a major role in monocyte function: p38MAPK, ERK and Akt. In addition, biological functions downstream of these signaling pathways were studied, including cytokine production, phagocytosis and differentiation into macrophages. To this end, blood samples from healthy volunteers were spiked with diverse concentrations of tacrolimus and MPA. Tacrolimus (200 ng/ml) inhibited phosphorylation of p38MAPK by 30% (mean) in CD14+ monocytes which was significantly less than in activated CD3+ T cells (max 60%; p < 0.05). This immunosuppressive agent also partly inhibited p-AKT (14%). MPA, at a therapeutic concentration showed the strongest effect on p-AKT (27% inhibition). p-ERK was inhibited with a maximum of 15% after spiking with either tacrolimus or MPA. The production of IL-1β and phagocytosis by monocytes were not affected by tacrolimus concentrations, whereas MPA did inhibit IL-1β production by 50%. Monocyte/macrophage polarization was shifted to an M2-like phenotype in the presence of tacrolimus, while MPA increased the expression of M2 surface markers, including CD163 and CD200R, on M1 macrophages. These results show that tacrolimus and MPA do not strongly affect monocyte function, apart from a change in macrophage polarization, to a clinically relevant degree.
Collapse
|
45
|
Leroi N, Lallemand F, Coucke P, Noel A, Martinive P. Impacts of Ionizing Radiation on the Different Compartments of the Tumor Microenvironment. Front Pharmacol 2016; 7:78. [PMID: 27064581 PMCID: PMC4811953 DOI: 10.3389/fphar.2016.00078] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/14/2016] [Indexed: 01/13/2023] Open
Abstract
Radiotherapy (RT) is one of the most important modalities for cancer treatment. For many years, the impact of RT on cancer cells has been extensively studied. Recently, the tumor microenvironment (TME) emerged as one of the key factors in therapy resistance. RT is known to influence and modify diverse components of the TME. Hence, we intent to review data from the literature on the impact of low and high single dose, as well as fractionated RT on host cells (endothelial cells, fibroblasts, immune and inflammatory cells) and the extracellular matrix. Optimizing the schedule of RT (i.e., dose per fraction) and other treatment modalities is a current challenge. A better understanding of the cascade of events and TME remodeling following RT would be helpful to design optimal treatment combination.
Collapse
Affiliation(s)
- Natacha Leroi
- Laboratory of Tumor and Development Biology, Groupe Interdisciplinaire de Génoprotéomique Appliquée-Cancer, University of Liège Liège, Belgium
| | - François Lallemand
- Laboratory of Tumor and Development Biology, Groupe Interdisciplinaire de Génoprotéomique Appliquée-Cancer, University of LiègeLiège, Belgium; Cyclotron Research Center, University of LiègeLiège, Belgium
| | - Philippe Coucke
- Radiotherapy-Oncology Department, Centre Hospitalier Universitaire de Liège Liège, Belgium
| | - Agnès Noel
- Laboratory of Tumor and Development Biology, Groupe Interdisciplinaire de Génoprotéomique Appliquée-Cancer, University of Liège Liège, Belgium
| | - Philippe Martinive
- Laboratory of Tumor and Development Biology, Groupe Interdisciplinaire de Génoprotéomique Appliquée-Cancer, University of LiègeLiège, Belgium; Radiotherapy-Oncology Department, Centre Hospitalier Universitaire de LiègeLiège, Belgium
| |
Collapse
|
46
|
Mavragani IV, Laskaratou DA, Frey B, Candéias SM, Gaipl US, Lumniczky K, Georgakilas AG. Key mechanisms involved in ionizing radiation-induced systemic effects. A current review. Toxicol Res (Camb) 2016; 5:12-33. [PMID: 30090323 PMCID: PMC6061884 DOI: 10.1039/c5tx00222b] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 08/06/2015] [Indexed: 12/11/2022] Open
Abstract
Organisms respond to physical, chemical and biological threats by a potent inflammatory response, aimed at preserving tissue integrity and restoring tissue homeostasis and function. Systemic effects in an organism refer to an effect or phenomenon which originates at a specific point and can spread throughout the body affecting a group of organs or tissues. Ionizing radiation (IR)-induced systemic effects arise usually from a local exposure of an organ or part of the body. This stress induces a variety of responses in the irradiated cells/tissues, initiated by the DNA damage response and DNA repair (DDR/R), apoptosis or immune response, including inflammation. Activation of this IR-response (IRR) system, especially at the organism level, consists of several subsystems and exerts a variety of targeted and non-targeted effects. Based on the above, we believe that in order to understand this complex response system better one should follow a 'holistic' approach including all possible mechanisms and at all organization levels. In this review, we describe the current status of knowledge on the topic, as well as the key molecules and main mechanisms involved in the 'spreading' of the message throughout the body or cells. Last but not least, we discuss the danger-signal mediated systemic immune effects of radiotherapy for the clinical setup.
Collapse
Affiliation(s)
- Ifigeneia V Mavragani
- Physics Department , School of Applied Mathematical and Physical Sciences , National Technical University of Athens (NTUA) , Zografou 15780 , Athens , Greece . ; ; Tel: +30-210-7724453
| | - Danae A Laskaratou
- Physics Department , School of Applied Mathematical and Physical Sciences , National Technical University of Athens (NTUA) , Zografou 15780 , Athens , Greece . ; ; Tel: +30-210-7724453
| | - Benjamin Frey
- Department of Radiation Oncology , University Hospital Erlangen , Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Serge M Candéias
- iRTSV-LCBM , CEA , Grenoble F-38000 , France
- IRTSV-LCBM , CNRS , Grenoble F-38000 , France
- iRTSV-LCBM , Univ. Grenoble Alpes , Grenoble F-38000 , France
| | - Udo S Gaipl
- Department of Radiation Oncology , University Hospital Erlangen , Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Katalin Lumniczky
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene , Budapest , Hungary
| | - Alexandros G Georgakilas
- Physics Department , School of Applied Mathematical and Physical Sciences , National Technical University of Athens (NTUA) , Zografou 15780 , Athens , Greece . ; ; Tel: +30-210-7724453
| |
Collapse
|
47
|
Kumari A, Simon SS, Moody TD, Garnett-Benson C. Immunomodulatory effects of radiation: what is next for cancer therapy? Future Oncol 2015; 12:239-56. [PMID: 26621553 DOI: 10.2217/fon.15.300] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Despite its former reputation as being immunosuppressive, it has become evident that radiation therapy can enhance antitumor immune responses. This quality can be harnessed by utilizing radiation as an adjuvant to cancer immunotherapies. Most studies combine the standard radiation dose and regimens indicated for the given disease state, with novel cancer immunotherapies. It has become apparent that low-dose radiation, as well as doses within the hypofractionated range, can modulate tumor cells making them better targets for immune cell reactivity. Herein, we describe the range of phenotypic changes induced in tumor cells by radiation, and explore the diverse mechanisms of immunogenic modulation reported at these doses. We also review the impact of these doses on the immune cell function of cytotoxic cells in vivo and in vitro.
Collapse
Affiliation(s)
- Anita Kumari
- Department of Biology, Georgia State University, 161 Jesse Hill Jr Dr, Atlanta, GA, USA
| | - Samantha S Simon
- Department of Biology, Georgia State University, 161 Jesse Hill Jr Dr, Atlanta, GA, USA
| | - Tomika D Moody
- Department of Biology, Georgia State University, 161 Jesse Hill Jr Dr, Atlanta, GA, USA
| | | |
Collapse
|
48
|
Modulation of inflammation by low and high doses of ionizing radiation: Implications for benign and malign diseases. Cancer Lett 2015; 368:230-7. [DOI: 10.1016/j.canlet.2015.04.010] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 12/31/2022]
|
49
|
Hellweg CE. The Nuclear Factor κB pathway: A link to the immune system in the radiation response. Cancer Lett 2015; 368:275-89. [DOI: 10.1016/j.canlet.2015.02.019] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/08/2015] [Accepted: 02/10/2015] [Indexed: 01/01/2023]
|
50
|
Reichl B, Block A, Schäfer U, Bert C, Müller R, Jung H, Rödel F. DEGRO practical guidelines for radiotherapy of non-malignant disorders. Strahlenther Onkol 2015; 191:701-9. [DOI: 10.1007/s00066-015-0865-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 06/03/2015] [Indexed: 11/29/2022]
|