1
|
Averbeck D. Low-Dose Non-Targeted Effects and Mitochondrial Control. Int J Mol Sci 2023; 24:11460. [PMID: 37511215 PMCID: PMC10380638 DOI: 10.3390/ijms241411460] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Non-targeted effects (NTE) have been generally regarded as a low-dose ionizing radiation (IR) phenomenon. Recently, regarding long distant abscopal effects have also been observed at high doses of IR) relevant to antitumor radiation therapy. IR is inducing NTE involving intracellular and extracellular signaling, which may lead to short-ranging bystander effects and distant long-ranging extracellular signaling abscopal effects. Internal and "spontaneous" cellular stress is mostly due to metabolic oxidative stress involving mitochondrial energy production (ATP) through oxidative phosphorylation and/or anaerobic pathways accompanied by the leakage of O2- and other radicals from mitochondria during normal or increased cellular energy requirements or to mitochondrial dysfunction. Among external stressors, ionizing radiation (IR) has been shown to very rapidly perturb mitochondrial functions, leading to increased energy supply demands and to ROS/NOS production. Depending on the dose, this affects all types of cell constituents, including DNA, RNA, amino acids, proteins, and membranes, perturbing normal inner cell organization and function, and forcing cells to reorganize the intracellular metabolism and the network of organelles. The reorganization implies intracellular cytoplasmic-nuclear shuttling of important proteins, activation of autophagy, and mitophagy, as well as induction of cell cycle arrest, DNA repair, apoptosis, and senescence. It also includes reprogramming of mitochondrial metabolism as well as genetic and epigenetic control of the expression of genes and proteins in order to ensure cell and tissue survival. At low doses of IR, directly irradiated cells may already exert non-targeted effects (NTE) involving the release of molecular mediators, such as radicals, cytokines, DNA fragments, small RNAs, and proteins (sometimes in the form of extracellular vehicles or exosomes), which can induce damage of unirradiated neighboring bystander or distant (abscopal) cells as well as immune responses. Such non-targeted effects (NTE) are contributing to low-dose phenomena, such as hormesis, adaptive responses, low-dose hypersensitivity, and genomic instability, and they are also promoting suppression and/or activation of immune cells. All of these are parts of the main defense systems of cells and tissues, including IR-induced innate and adaptive immune responses. The present review is focused on the prominent role of mitochondria in these processes, which are determinants of cell survival and anti-tumor RT.
Collapse
Affiliation(s)
- Dietrich Averbeck
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France
| |
Collapse
|
2
|
Gao L, Zhang A. Low-dose radiotherapy effects the progression of anti-tumor response. Transl Oncol 2023; 35:101710. [PMID: 37320873 DOI: 10.1016/j.tranon.2023.101710] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 06/17/2023] Open
Abstract
The history of low-dose radiotherapy (LDRT or LDR) as a treatment modality for malignant tumors dates back to the 1920s. Even with the minimal total dose administered during treatment, LDRT can result in long-lasting remission. Autocrine and paracrine signaling are widely recognized for fostering the growth and development of tumor cells. LDRT exerts systemic anti-tumor effects through various mechanisms, such as enhancing the activity of immune cells and cytokines, shifting the immune response towards an anti-tumor phenotype, influencing gene expression, and blocking crucial immunosuppressive pathways. Additionally, LDRT has been demonstrated to enhance the infiltration of activated T cells and initiate a series of inflammatory processes while modulating the tumor microenvironment. In this context, the objective of receiving radiation is not to directly kill tumor cells but to reprogram the immune system. Enhancing anti-tumor immunity may be a critical mechanism by which LDRT plays a role in cancer suppression. Therefore, this review primarily focuses on the clinical and preclinical efficacy of LDRT in combination with other anti-cancer strategies, such as the interaction between LDRT and the tumor microenvironment, and the remodeling of the immune system.
Collapse
Affiliation(s)
- Lei Gao
- Medical Imaging Department, Huabei Petroleum Administration Bureau General Hospital, Renqiu, China
| | - Anqi Zhang
- Oncology Department, Huabei Petroleum Administration Bureau General Hospital, Renqiu, China.
| |
Collapse
|
3
|
Wang J, Zhang J, Wen W, Wang F, Wu M, Chen D, Yu J. Exploring low-dose radiotherapy to overcome radio-immunotherapy resistance. Biochim Biophys Acta Mol Basis Dis 2023:166789. [PMID: 37302425 DOI: 10.1016/j.bbadis.2023.166789] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/13/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the current treatment landscape for cancer, yet the response rates of ICIs remain unmet. Synergistic with immunotherapy, low-dose radiotherapy (LDRT) has been demonstrated to activate anti-tumor immunity - a transition from traditional radiation therapy geared toward local radical treatment to a type of immunological adjuvant. As such, studies utilizing LDRT to enhance the efficacy of immunotherapy have been increasing preclinically and clinically. This paper reviews the recent strategies of using LDRT to overcome the resistance of ICIs, as well as providing potential opportunities in cancer treatment. Despite the potential of LDRT in immunotherapy is recognized, the mechanisms behind this form of treatment remain largely elusive. Thus, we reviewed history, mechanisms and challenges associated with this form of treatment, as well as different modes of its application, to provide relatively accurate practice standards for LDRT as a sensitizing treatment when combined with immunotherapy or radio-immunotherapy.
Collapse
Affiliation(s)
- Juan Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Jingxin Zhang
- Shandong University Cancer Center, Jinan, Shandong 250117, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Weitao Wen
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Fei Wang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Meng Wu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Dawei Chen
- Shandong University Cancer Center, Jinan, Shandong 250117, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China.
| | - Jinming Yu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China; Shandong University Cancer Center, Jinan, Shandong 250117, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong 250117, PR China.
| |
Collapse
|
4
|
Laurent PA, Morel D, Meziani L, Depil S, Deutsch E. Radiotherapy as a means to increase the efficacy of T-cell therapy in solid tumors. Oncoimmunology 2022; 12:2158013. [PMID: 36567802 PMCID: PMC9788698 DOI: 10.1080/2162402x.2022.2158013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cells have demonstrated significant improvements in the treatment of refractory B-cell malignancies that previously showed limited survival. In contrast, early-phase clinical studies targeting solid tumors have been disappointing. This may be due to both a lack of specific and homogeneously expressed targets at the surface of tumor cells, as well as intrinsic properties of the solid tumor microenvironment that limit homing and activation of adoptive T cells. Faced with these antagonistic conditions, radiotherapy (RT) has the potential to change the overall tumor landscape, from depleting tumor cells to reshaping the tumor microenvironment. In this article, we describe the current landscape and discuss how RT may play a pivotal role for enhancing the efficacy of adoptive T-cell therapies in solid tumors. Indeed, by improving homing, expansion and activation of infused T cells while reducing tumor volume and heterogeneity, the use of RT could help the implementation of engineered T cells in the treatment of solid tumors.
Collapse
Affiliation(s)
- Pierre-Antoine Laurent
- Department of Radiation Oncology, Gustave Roussy Cancer Campus; UNICANCER, Villejuif, France
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, University of Paris-Saclay, SIRIC SOCRATE, Villejuif, France
| | - Daphne Morel
- Drug Development Department (D.I.T.E.P), Gustave Roussy Cancer Campus; UNICANCER, Villejuif, France
| | - Lydia Meziani
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, University of Paris-Saclay, SIRIC SOCRATE, Villejuif, France
| | | | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy Cancer Campus; UNICANCER, Villejuif, France
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, University of Paris-Saclay, SIRIC SOCRATE, Villejuif, France
| |
Collapse
|
5
|
Nagpal I, Yuan ZM. p53-mediated metabolic response to low doses of ionizing radiation. Int J Radiat Biol 2022; 99:934-940. [DOI: 10.1080/09553002.2022.2142983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Isha Nagpal
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zhi-Min Yuan
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
6
|
Wang XC, Tian LL, Fan CX, Duo CH, Xu KM. The Adaptive Responses in Non-Small Cell Lung Cancer A549 Cell Lines Induced by Low-Dose Ionizing Radiation and the Variations of miRNA Expression. Dose Response 2021; 19:15593258211039931. [PMID: 34658683 PMCID: PMC8516394 DOI: 10.1177/15593258211039931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/29/2021] [Indexed: 11/17/2022] Open
Abstract
Objective To study the effects of adaptive response in A549 cells induced by low-dose radiation and the miRNAs expression. Methods A549 cells were irradiated with 50 mGy and 200 mGy initial doses, respectively, and then irradiated with a challenge dose 20 Gy at 6 hours interval. The biological effects and miRNA expression were detected. Results The apoptosis rates of 50 mGy-20 Gy and 200 mGy-20 Gy groups were significantly lower than that of only 20 Gy irradiation group (P < .05). The percentage of G2/M phase cells of 50 mGy-20 Gy and 200 mGy-20 Gy groups was significantly decreased relative to the 20 Gy group (P < .05). One miRNA (mir-3662) was upregulated and 15 miRNAs (mir-185, mir-1908, mir-307, mir-182, mir-92a, mir-582, mi-r501, mir138-5p, mir-1260, mir-484, mir-378d, mir-193b, mir-127-3p, mir-1303, and mir-654-5p) were downregulated both in 50 mGy-20 Gy and 200 mGy-20 Gy groups than that of the 20 Gy group. Go and KEGG enrichment analysis showed that the target genes were significantly enriched in cell communication regulation, metabolic process, enzyme binding, and catalytic activity signaling pathways. Conclusion Low-dose X-ray of 50 mGy and 200 mGy radiation can induce adaptive apoptosis response prior to 20 Gy in A549 cells. Sixteen differently expressed miRNAs may play important roles in the adaptive effect of low-dose radiation.
Collapse
Affiliation(s)
- Xiao-Chun Wang
- ShiLong Hospital (Research Center for Pneumoconiosis Prevention and Treatment), National Center for Occupational Safety and Health, NHC, Beijing, China
| | - Li-Li Tian
- nstitute of Infectious and Endemic Disease Prevention, Beijing Centers for Disease Control and Prevention, Beijing, China
| | | | - Cai-Hong Duo
- ShiLong Hospital (Research Center for Pneumoconiosis Prevention and Treatment), National Center for Occupational Safety and Health, NHC, Beijing, China
| | - Ke-Ming Xu
- ShiLong Hospital (Research Center for Pneumoconiosis Prevention and Treatment), National Center for Occupational Safety and Health, NHC, Beijing, China
| |
Collapse
|
7
|
Dawood A, Mothersill C, Seymour C. Low dose ionizing radiation and the immune response: what is the role of non-targeted effects? Int J Radiat Biol 2021; 97:1368-1382. [PMID: 34330196 DOI: 10.1080/09553002.2021.1962572] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES This review aims to trace the historical narrative surrounding the low dose effects of radiation on the immune system and how our understanding has changed from the beginning of the 20th century to now. The particular focus is on the non-targeted effects (NTEs) of low dose ionizing radiation (LDIR) which are effects that occur when irradiated cells emit signals that cause effects in the nearby or distant non-irradiated cells known as radiation induced bystander effect (RIBE). Moreover, radiation induced genomic instability (RIGI) and abscopal effect (AE) also regarded as NTE. This was prompted by our recent discovery that ultraviolet A (UVA) photons are emitted by the irradiated cells and that these photons can trigger NTE such as the RIBE in unirradiated recipients of these photons. Given the well-known association between UV radiation and the immune response, where these biophotons may pose as bystander signals potentiating processes in deep tissues as a consequence of LDIR, it is timely to review the field with a fresh lens. Various pathways and immune components that contribute to the beneficial and adverse types of modulation induced by LDR will also be revisited. CONCLUSION There is limited evidence for LDIR induced immune effects by way of a non-targeted mechanism in biological tissue. The literature examining low to medium dose effects of ionizing radiation on the immune system and its components is complex and controversial. Early work was compromised by lack of good dosimetry while later work mainly looks at the involvement of immune response in radiotherapy. There is a lack of research in the LDIR/NTE field focusing on immune response although bone marrow stem cells and lineages were critical in the identification and characterization of NTE where effects like RIGI and RIBE were heavily researched. This may be in part, a result of the difficulty of isolating NTE in whole organisms which are essential for good immune response studies. Models involving inter organism transmission of NTE are a promising route to overcome these issues.
Collapse
Affiliation(s)
- Annum Dawood
- Department of Physics and Astronomy, McMaster University, Hamilton, Canada
| | | | - Colin Seymour
- Department of Biology, McMaster University, Hamilton, Canada
| |
Collapse
|
8
|
Nowosielska EM, Cheda A, Pociegiel M, Cheda L, Szymański P, Wiedlocha A. Effects of a Unique Combination of the Whole-Body Low Dose Radiotherapy with Inactivation of Two Immune Checkpoints and/or a Heat Shock Protein on the Transplantable Lung Cancer in Mice. Int J Mol Sci 2021; 22:6309. [PMID: 34208396 PMCID: PMC8231142 DOI: 10.3390/ijms22126309] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 11/25/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) continues to be the leading cause of cancer death worldwide. Recently, targeting molecules whose functions are associated with tumorigenesis has become a game changing adjunct to standard anti-cancer therapy. As evidenced by the results of preclinical and clinical investigations, whole-body irradiations (WBI) with X-rays at less than 0.1-0.2 Gy per fraction can induce remissions of various neoplasms without inciting adverse side effects of conventional chemo- and radiotherapy. In the present study, a murine model of human NSCLC was employed to evaluate for the first time the anti-neoplastic efficacy of WBI combined with inactivation of CTLA-4, PD-1, and/or HSP90. The results indicate that WBI alone and in conjunction with the inhibition of the function of the cytotoxic T-lymphocyte antigen-4 (CTLA-4) and the programmed death-1 (PD-1) receptor immune checkpoints (ICs) and/or heat shock protein 90 (HSP90) markedly reduced tumorigenesis in mice implanted by three different routes with the syngeneic Lewis lung cancer cells and suppressed clonogenic potential of Lewis lung carcinoma (LLC1) cells in vitro. These results were associated with the relevant changes in the profile of pro- and anti-neoplastic immune cells recruited to the growing tumors and the circulating anti- and pro-inflammatory cytokines. In contrast, inhibition of the tested molecular targets used either separately or in combination with each other did not exert notable anti-neoplastic effects. Moreover, no significant synergistic effects were detected when the inhibitors were applied concurrently with WBI. The obtained results supplemented with further mechanistic explanations provided by future investigations will help design the effective strategies of treatment of lung and other cancers based on inactivation of the immune checkpoint and/or heat shock molecules combined with low-dose radiotherapy.
Collapse
Affiliation(s)
- Ewa M. Nowosielska
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163 Warsaw, Poland; (A.C.); (P.S.); (A.W.)
| | - Aneta Cheda
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163 Warsaw, Poland; (A.C.); (P.S.); (A.W.)
| | - Mateusz Pociegiel
- National Centre for Nuclear Research Radioisotope Centre POLATOM, 7A Soltana St., 05-400 Otwock, Poland;
| | - Lukasz Cheda
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, 101 Żwirki i Wigury St., 02-089 Warsaw, Poland;
| | - Paweł Szymański
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163 Warsaw, Poland; (A.C.); (P.S.); (A.W.)
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, 1 Muszyńskiego St., 90-151 Lodz, Poland
| | - Antoni Wiedlocha
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163 Warsaw, Poland; (A.C.); (P.S.); (A.W.)
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway
- Centre for Cancer Reprograming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
| |
Collapse
|
9
|
McCarron RA, Barnard SGR, Babini G, Dalke C, Graw J, Leonardi S, Mancuso M, Moquet JE, Pawliczek D, Pazzaglia S, De Stefano I, Ainsbury EA. Radiation-Induced Lens Opacity and Cataractogenesis: A Lifetime Study Using Mice of Varying Genetic Backgrounds. Radiat Res 2021; 197:57-66. [PMID: 33984859 DOI: 10.1667/rade-20-00266.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/20/2021] [Indexed: 11/03/2022]
Abstract
Recent epidemiological findings and reanalysis of historical data suggest lens opacities resulting from ionizing radiation exposures are likely induced at lower doses than previously thought. These observations have led to ICRP recommendations for a reduction in the occupational dose limits for the eye lens, as well as subsequent implementation in EU member states. The EU CONCERT LDLensRad project was initiated to further understand the effects of ionizing radiation on the lens and identify the mechanism(s) involved in radiation-induced cataract, as well as the impact of dose and dose-rate. Here, we present the results of a long-term study of changes to lens opacity in male and female adult mice from a variety of different genetic (radiosensitive or radioresistant) backgrounds, including mutant strains Ercc2 and Ptch1, which were assumed to be susceptible to radiation-induced lens opacities. Mice received 0.5, 1 and 2 Gy 60Co gamma-ray irradiation at dose rates of 0.063 and 0.3 Gy min-1. Scheimpflug imaging was used to quantify lens opacification as an early indicator of cataract, with monthly observations taken postirradiation for an 18-month period in all strains apart from 129S2, which were observed for 12 months. Opacification of the lens was found to increase with time postirradiation (with age) for most mouse models, with ionizing radiation exposure increasing opacities further. Sex, dose, dose rate and genetic background were all found to be significant contributors to opacification; however, significant interactions were identified, which meant that the impact of these factors was strain dependent. Mean lens density increased with higher dose and dose rate in the presence of Ercc2 and Ptch1 mutations. This project was the first to focus on low (<1 Gy) dose, multiple dose rate, sex and strain effects in lens opacification, and clearly demonstrates the importance of these experimental factors in radiobiological investigations on the lens. The results provide insight into the effects of ionizing radiation on the lens as well as the need for further work in this area to underpin appropriate radiation protection legislation and guidance.
Collapse
Affiliation(s)
- R A McCarron
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, United Kingdom
| | - S G R Barnard
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, United Kingdom.,Durham University, School of Biosciences, Durham, United Kingdom
| | - G Babini
- Department of Physics, University of Pavia, Pavia, Italy.,Department of Woman and Child Health, Fondazione Policlinico A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - C Dalke
- Helmholtz Zentrum München GmbH, German Research Centre for Environmental Health, Neuherberg, Germany
| | - J Graw
- Helmholtz Zentrum München GmbH, German Research Centre for Environmental Health, Neuherberg, Germany
| | - S Leonardi
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - M Mancuso
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - J E Moquet
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, United Kingdom
| | - D Pawliczek
- Helmholtz Zentrum München GmbH, German Research Centre for Environmental Health, Neuherberg, Germany
| | - S Pazzaglia
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - I De Stefano
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - E A Ainsbury
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, United Kingdom
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Deana Y, Burgara-Estrella AJ, Montalvo-Corral M, Angulo-Molina A, Acosta-Elías MA, Silva-Campa E, Sarabia-Sainz JA, Rodríguez-Hernández IC, Pedroza-Montero MR. Effect of gamma irradiation doses in the structural and functional properties of mice splenic cells. Int J Radiat Biol 2018; 95:286-297. [PMID: 30496016 DOI: 10.1080/09553002.2019.1547435] [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: 10/27/2022]
Abstract
PURPOSE Ionizing radiation is nowadays effectively used in cancer treatments. However, the effect of irradiation in immune-system cells is poorly understood and remains controversial. The aim of this work was to determine the effect of γ-irradiation in the structural and functional properties of mice splenic cells. MATERIALS AND METHODS Structural traits of irradiated splenic cells were evaluated by Atomic Force Microscopy and Raman spectroscopy. Functional properties were measured by gene and protein expression by RT-qPCR and ELISA, respectively. The induced cytotoxic effect was evaluated by MTT assay and the phagocytic capability by flow cytometry. RESULTS Membrane roughness and molecular composition of splenic adherent cells are not changed by irradiation doses exposure. An increase in transcription of pro-inflammatory cytokines was observed. While protein expression decreased in IL-2 dose-dependent, relevant differences were identified in the anti-inflammatory marker IL-10 at 27 Gy. An increase of cytotoxicity in irradiated cells at 7 Gy and 27 Gy doses was observed, while phagocytosis was slight increased at 7 Gy dose but not statistically significant. CONCLUSIONS We have demonstrated that γ-irradiation affects the splenic cells and changes the cytokines profile toward a pro-inflammatory phenotype and a tendency to increase the cytotoxicity was found, which implies a stimulation of immune response induced by γ-irradiation.
Collapse
Affiliation(s)
- Yanik Deana
- a Departamento de Investigación en Física , Universidad de Sonora , Hermosillo , México.,b Institute for Chemistry and Bioanalytics , University of Applied Sciences and Arts Northwestern , Muttenz , Switzerland
| | | | - Maricela Montalvo-Corral
- c Departamento de Nutrición , Centro de Investigación en Alimentación y Desarrollo A.C. , Hermosillo , México
| | | | - Mónica A Acosta-Elías
- a Departamento de Investigación en Física , Universidad de Sonora , Hermosillo , México
| | - Erika Silva-Campa
- a Departamento de Investigación en Física , Universidad de Sonora , Hermosillo , México
| | - Jose A Sarabia-Sainz
- a Departamento de Investigación en Física , Universidad de Sonora , Hermosillo , México
| | | | | |
Collapse
|
13
|
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]
|
14
|
Nowosielska EM, Cheda A, Zdanowski R, Lewicki S, Scott BR, Janiak MK. Effect of internal contamination with tritiated water on the neoplastic colonies in the lungs, innate anti-tumour reactions, cytokine profile, and haematopoietic system in radioresistant and radiosensitive mice. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2018; 57:251-264. [PMID: 29626227 PMCID: PMC6060768 DOI: 10.1007/s00411-018-0739-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/21/2018] [Indexed: 05/14/2023]
Abstract
Tritium is a potentially significant source of internal radiation exposure which, at high levels, can be carcinogenic. We evaluated whether single intraperitoneal injection of BALB/c and C57BL/6 mice with tritiated water (HTO) leading to exposure to low (0.01 or 0.1 Gy) and intermediate (1.0 Gy) cumulative whole-body doses of β radiation is immunosuppressive, as judged by enhancement of artificial tumour metastases, functioning of NK lymphocytes and macrophages, circulating cytokine's levels, and numbers of bone marrow, spleen, and peripheral blood cells. We demonstrate that internal contamination of radiosensitive BALB/c and radioresistant C57BL/6 mice with HTO at all the absorbed doses tested did not affect the development of neoplastic colonies in the lungs caused by intravenous injection of syngeneic cancer cells. However, internal exposure of BALB/c and C57BL/6 mice to 0.1 and 0.01 Gy of β radiation, respectively, up-regulated cytotoxic activity of and IFN-γ synthesis in NK lymphocytes and boosted macrophage secretion of nitric oxide. Internal contamination with HTO did not affect the serum levels of pro- (IL-1β, IL-2, IL-6, TNF-α,) and anti-inflammatory (IL-1Ra, IL-4, IL-10) cytokines. In addition, exposure of mice of both strains to low and intermediate doses from the tritium-emitted β-particles did not result in any significant changes in the numbers of bone marrow, spleen, and peripheral blood cells. Overall, our data indicate that internal tritium contamination of both radiosensitive and radioresistant mice leading to low and intermediate absorbed β-radiation doses is not immunosuppressive but may enhance some but not all components of anticancer immunity.
Collapse
Affiliation(s)
- Ewa M Nowosielska
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland.
| | - Aneta Cheda
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| | - Robert Zdanowski
- Department of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| | - Sławomir Lewicki
- Department of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| | - Bobby R Scott
- Lovelace Respiratory Research Institute, 2425 Ridgecrest Drive SE, Albuquerque, 87108, NM, USA
| | - Marek K Janiak
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| |
Collapse
|
15
|
Vaiserman A, Koliada A, Zabuga O, Socol Y. Health Impacts of Low-Dose Ionizing Radiation: Current Scientific Debates and Regulatory Issues. Dose Response 2018; 16:1559325818796331. [PMID: 30263019 PMCID: PMC6149023 DOI: 10.1177/1559325818796331] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 12/31/2022] Open
Abstract
Health impacts of low-dose ionizing radiation are significant in important fields such as X-ray imaging, radiation therapy, nuclear power, and others. However, all existing and potential applications are currently challenged by public concerns and regulatory restrictions. We aimed to assess the validity of the linear no-threshold (LNT) model of radiation damage, which is the basis of current regulation, and to assess the justification for this regulation. We have conducted an extensive search in PubMed. Special attention has been given to papers cited in comprehensive reviews of the United States (2006) and French (2005) Academies of Sciences and in the United Nations Scientific Committee on Atomic Radiation 2016 report. Epidemiological data provide essentially no evidence for detrimental health effects below 100 mSv, and several studies suggest beneficial (hormetic) effects. Equally significant, many studies with in vitro and in animal models demonstrate that several mechanisms initiated by low-dose radiation have beneficial effects. Overall, although probably not yet proven to be untrue, LNT has certainly not been proven to be true. At this point, taking into account the high price tag (in both economic and human terms) borne by the LNT-inspired regulation, there is little doubt that the present regulatory burden should be reduced.
Collapse
|
16
|
Proton irradiation orchestrates macrophage reprogramming through NFκB signaling. Cell Death Dis 2018; 9:728. [PMID: 29950610 PMCID: PMC6021396 DOI: 10.1038/s41419-018-0757-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/27/2018] [Accepted: 05/29/2018] [Indexed: 12/28/2022]
Abstract
Tumor-associated macrophages (TAMs) represent potential targets for anticancer treatments as these cells play critical roles in tumor progression and frequently antagonize the response to treatments. TAMs are usually associated to an M2-like phenotype, characterized by anti-inflammatory and protumoral properties. This phenotype contrasts with the M1-like macrophages, which exhibits proinflammatory, phagocytic, and antitumoral functions. As macrophages hold a high plasticity, strategies to orchestrate the reprogramming of M2-like TAMs towards a M1 antitumor phenotype offer potential therapeutic benefits. One of the most used anticancer treatments is the conventional X-ray radiotherapy (RT), but this therapy failed to reprogram TAMs towards an M1 phenotype. While protontherapy is more and more used in clinic to circumvent the side effects of conventional RT, the effects of proton irradiation on macrophages have not been investigated yet. Here we showed that M1 macrophages (THP-1 cell line) were more resistant to proton irradiation than unpolarized (M0) and M2 macrophages, which correlated with differential DNA damage detection. Moreover, proton irradiation-induced macrophage reprogramming from M2 to a mixed M1/M2 phenotype. This reprogramming required the nuclear translocation of NFκB p65 subunit as the inhibition of IκBα phosphorylation completely reverted the macrophage re-education. Altogether, the results suggest that proton irradiation promotes NFκB-mediated macrophage polarization towards M1 and opens new perspectives for macrophage targeting with charged particle therapy.
Collapse
|
17
|
Shimura N, Kojima S. The Lowest Radiation Dose Having Molecular Changes in the Living Body. Dose Response 2018; 16:1559325818777326. [PMID: 29977175 PMCID: PMC6024299 DOI: 10.1177/1559325818777326] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/22/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022] Open
Abstract
We herein attempted to identify the lowest radiation dose causing molecular changes in the living body. We investigated the effects of radiation in human cells, animals, and humans. DNA double-strand breaks (DSBs) formed in cells at γ- or X-ray irradiation doses between 1 mGy and 0.5 Gy; however, the extent of DSB formation differed depending on the cell species. The formation of micronuclei (MNs) and nucleoplasmic bridges (NPBs) was noted at radiation doses between 0.1 and 0.2 Gy. Stress-responsive genes were upregulated by lower radiation doses than those that induced DNA DSBs or MN and NPBs. These γ- or X-ray radiation doses ranged between approximately 10 and 50 mGy. In animals, chromosomal aberrations were detected between 50 mGy and 0.1 Gy of low linear energy transfer radiation, 0.1 Gy of metal ion beams, and 9 mGy of fast neutrons. In humans, DNA damage has been observed in children who underwent computed tomography scans with an estimated blood radiation dose as low as 0.15 mGy shortly after examination. The frequencies of chromosomal translocations were lower in residents of high background areas than in those of control areas. In humans, systemic adaptive responses may have been prominently expressed at these radiation doses.
Collapse
Affiliation(s)
- Noriko Shimura
- Faculty of Pharmaceutical Sciences, Ohu University, Tomita-machi, Koriyama, Fukushima, Japan
| | - Shuji Kojima
- Faculty of Pharmaceutical Sciences, Department of Radiation Biosciences, Tokyo University of Science (TUS), Chiba, Japan
| |
Collapse
|
18
|
Kandil EI, El-Sonbaty SM, Moawed FS, Khedr OM. Anticancer redox activity of gallium nanoparticles accompanied with low dose of gamma radiation in female mice. Tumour Biol 2018; 40:1010428317749676. [PMID: 29587600 DOI: 10.1177/1010428317749676] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Guided treatments with nanoparticles and radiotherapy are a new approach in cancer therapy. This study evaluated the beneficial antitumor effects of γ-radiation together with gallium nanoparticles against solid Ehrlich carcinoma in female mice. Gallium nanoparticles were biologically synthesized using Lactobacillus helveticus cells. Transmission electron microscopy showed gallium nanoparticles with size range of 8-20 nm. In vitro study of gallium nanoparticles on MCF-7 revealed IC50 of 8.0 μg. Gallium nanoparticles (0.1 mg/kg body weight) were injected intraperitoneally daily on the seventh day of Ehrlich carcinoma cells inoculation. Whole-body γ-radiation was carried out at a single dose of 0.25 Gy on eighth day after tumor inoculation. Biochemical analysis showed that solid Ehrlich carcinoma induced a significant increase in alanine aminotransferase activity and creatinine level in serum, calcium, and iron concentrations in liver tissue compared to normal control. Treatment of Ehrlich carcinoma-bearing mice with gallium nanoparticles and/or low dose of γ-radiation exposure significantly reduced tumor volume, decreased alanine aminotransferase and creatinine levels in serum, increased lipid peroxidation, and decreased glutathione content as well as calcium and iron concentrations in liver and tumor tissues with intense DNA fragmentation accompanied compared to untreated tumor cells. Moreover, mitochondria in the treated groups displayed a significant increase in Na+/K+-ATPase, complexes II and III with significant reduction in CYP450 gene expression, which may indicate a synergistic effect of gallium nanoparticles and/or low dose of γ-radiation combination against Ehrlich carcinoma injury, and this results were well appreciated with the histopathological findings in the tumor tissue. We conclude that combined treatment of gallium nanoparticles and low dose of gamma-radiation resulted in suppressive induction of cytotoxic effects on cancer cells.
Collapse
Affiliation(s)
- Eman I Kandil
- 1 Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Sawsan M El-Sonbaty
- 2 Radiation Microbiology Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Fatma Sm Moawed
- 3 Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Ola Ms Khedr
- 1 Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Janiak MK, Wincenciak M, Cheda A, Nowosielska EM, Calabrese EJ. Cancer immunotherapy: how low-level ionizing radiation can play a key role. Cancer Immunol Immunother 2017; 66:819-832. [PMID: 28361232 PMCID: PMC5489643 DOI: 10.1007/s00262-017-1993-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 03/22/2017] [Indexed: 12/17/2022]
Abstract
The cancer immunoediting hypothesis assumes that the immune system guards the host against the incipient cancer, but also "edits" the immunogenicity of surviving neoplastic cells and supports remodeling of tumor microenvironment towards an immunosuppressive and pro-neoplastic state. Local irradiation of tumors during standard radiotherapy, by killing neoplastic cells and generating inflammation, stimulates anti-cancer immunity and/or partially reverses cancer-promoting immunosuppression. These effects are induced by moderate (0.1-2.0 Gy) or high (>2 Gy) doses of ionizing radiation which can also harm normal tissues, impede immune functions, and increase the risk of secondary neoplasms. In contrast, such complications do not occur with exposures to low doses (≤0.1 Gy for acute irradiation or ≤0.1 mGy/min dose rate for chronic exposures) of low-LET ionizing radiation. Furthermore, considerable evidence indicates that such low-level radiation (LLR) exposures retard the development of neoplasms in humans and experimental animals. Here, we review immunosuppressive mechanisms induced by growing tumors as well as immunomodulatory effects of LLR evidently or likely associated with cancer-inhibiting outcomes of such exposures. We also offer suggestions how LLR may restore and/or stimulate effective anti-tumor immunity during the more advanced stages of carcinogenesis. We postulate that, based on epidemiological and experimental data amassed over the last few decades, whole- or half-body irradiations with LLR should be systematically examined for its potential to be a viable immunotherapeutic treatment option for patients with systemic cancer.
Collapse
Affiliation(s)
- Marek K Janiak
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland.
| | - Marta Wincenciak
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| | - Aneta Cheda
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| | - Ewa M Nowosielska
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| | - Edward J Calabrese
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
22
|
Wainwright DA, Horbinski CM, Hashizume R, James CD. Therapeutic Hypothesis Testing With Rodent Brain Tumor Models. Neurotherapeutics 2017; 14:385-392. [PMID: 28321824 PMCID: PMC5398994 DOI: 10.1007/s13311-017-0523-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The development and application of rodent models for preclinical testing of novel therapeutics and approaches for treating brain tumors has been a mainstay of neuro-oncology preclinical research for decades, and is likely to remain so into the foreseeable future. These models serve as an important point of entry for analyzing the potential efficacy of experimental therapies that are being considered for clinical trial evaluation. Although rodent brain tumor models have seen substantial change, particularly since the introduction of genetically engineered mouse models, certain principles associated with the use of these models for therapeutic testing are enduring, and form the basis for this review. Here we discuss the most common rodent brain tumor models while directing specific attention to their usefulness in preclinical evaluation of experimental therapies. These models include genetically engineered mice that spontaneously or inducibly develop brain tumors; syngeneic rodent models in which cultured tumor cells are engrafted into the same strain of rodent from which they were derived; and patient-derived xenograft models in which human tumor cells are engrafted in immunocompromised rodents. The emphasis of this review is directed to the latter.
Collapse
Affiliation(s)
- Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine-Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Craig M Horbinski
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rintaro Hashizume
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - C David James
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
23
|
Prakash H, Klug F, Nadella V, Mazumdar V, Schmitz-Winnenthal H, Umansky L. Low doses of gamma irradiation potentially modifies immunosuppressive tumor microenvironment by retuning tumor-associated macrophages: lesson from insulinoma. Carcinogenesis 2016; 37:301-313. [PMID: 26785731 DOI: 10.1093/carcin/bgw007] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 01/12/2016] [Indexed: 01/01/2023] Open
Abstract
Tumor infiltrating iNOS+ macrophages under the influence of immunosuppressive tumor microenvironment gets polarized to tumor-promoting and immunosuppressive macrophages, known as tumor-associated macrophages (TAM). Their recruitment and increased density in the plethora of tumors has been associated with poor prognosis in cancer patients. Therefore, retuning of TAM to M1 phenotype would be a key for effective immunotherapy. Radiotherapy has been a potential non-invasive strategy to improve cancer immunotherapy and tumor immune rejection. Irradiation of late-stage tumor-bearing Rip1-Tag5 mice twice with 2 Gy dose resulted in profound changes in the inflammatory tumor micromilieu, characterized by induction of M1-associated effecter cytokines as well as reduction in protumorigenic and M2-associated effecter cytokines. Similarly, in vitro irradiation of macrophages with 2 Gy dose-induced expression of iNOS, NO, NFκBpp65, pSTAT3 and proinflammatory cytokines secretion while downregulating p38MAPK which are involved in iNOS translation and acquisition of an M1-like phenotype. Enhancement of various M2 effecter cytokines and angiogenic reprogramming in iNOs+ macrophage depleted tumors and their subsequent reduction by 2 Gy dose in Rip1-Tag5 transgenic mice furthermore demonstrated a critical role of peritumoral macrophages in the course of gamma irradiation mediated M1 retuning of insulinoma.
Collapse
Affiliation(s)
- Hridayesh Prakash
- Translational Immunology Division , German Cancer Research Center (DKFZ) and National Center of Tumor Diseases (NCT) , Im-Neuenheimer Feld 460 , 69120 Heidelberg , Germany
| | - Felix Klug
- Translational Immunology Division , German Cancer Research Center (DKFZ) and National Center of Tumor Diseases (NCT) , Im-Neuenheimer Feld 460 , 69120 Heidelberg , Germany
| | - Vinod Nadella
- School of Life Sciences , University of Hyderabad , Hyderabad 500046 , India
| | - Varadendra Mazumdar
- School of Life Sciences , University of Hyderabad , Hyderabad 500046 , India
| | | | - Liudmila Umansky
- Translational Immunology Division , German Cancer Research Center (DKFZ) and National Center of Tumor Diseases (NCT) , Im-Neuenheimer Feld 460 , 69120 Heidelberg , Germany
| |
Collapse
|
24
|
The effect of radiation on the immune response to cancers. Int J Mol Sci 2014; 15:927-43. [PMID: 24434638 PMCID: PMC3907847 DOI: 10.3390/ijms15010927] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 12/26/2013] [Accepted: 12/31/2013] [Indexed: 02/07/2023] Open
Abstract
In cancer patients undergoing radiation therapy, the beneficial effects of radiation can extend beyond direct cytotoxicity to tumor cells. Delivery of localized radiation to tumors often leads to systemic responses at distant sites, a phenomenon known as the abscopal effect which has been attributed to the induction and enhancement of the endogenous anti-tumor innate and adaptive immune response. The mechanisms surrounding the abscopal effect are diverse and include trafficking of lymphocytes into the tumor microenvironment, enhanced tumor recognition and killing via up-regulation of tumor antigens and antigen presenting machinery and, induction of positive immunomodulatory pathways. Here, we discuss potential mechanisms of radiation-induced enhancement of the anti-tumor response through its effect on the host immune system and explore potential combinational immune-based strategies such as adoptive cellular therapy using ex vivo expanded NK and T cells as a means of delivering a potent effector population in the context of radiation-enhanced anti-tumor immune environment.
Collapse
|
25
|
Abstract
“At the early stages of evolution, increasingly complex organisms developed powerful defense mechanisms against such adverse radiation effects as mutation and malignant change. These effects originate in the cell nucleus, where the DNA is their primary target. That evolution has apparently proceeded for so long is proof, in part, of the effectiveness of living things’ defenses against radiation.” Zbigniew Jaworowski 1999 “Chernobyl was indeed an historic event; it is the only nuclear power station disaster that ever resulted in an occupational death toll, albeit a comparatively small one. A vast environmental dispersion of radioactivity occurred that did not cause any scientifically confirmed fatalities in the general population. The worst harm to the population was caused not by radiation, and not to flesh, but to minds.” Zbigniew Jaworowski 2010
Collapse
|
26
|
Scott BR, Bruce VR, Gott KM, Wilder J, March T. Small γ-Ray Doses Prevent Rather than Increase Lung Tumors in Mice. Dose Response 2012; 10:527-40. [PMID: 23304103 DOI: 10.2203/dose-response.12-035.scott] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We show evidence for low doses of γ rays preventing spontaneous hyperplastic foci and adenomas in the lungs of mice, presumably via activating natural anticancer defenses. The evidence partly relates to a new study we conducted whereby a small number of female A/J mice received 6 biweekly dose fractions (100 mGy per fraction) of γ rays to the total body which prevented the occurrence of spontaneous hyperplastic foci in the lung. We also analyzed data from a much earlier Oak Ridge National Laboratory study involving more than 10,000 female RFMf/Un mice whereby single γ-ray doses from 100 to 1,000 mGy prevented spontaneous lung adenomas. We point out the possibility that the decrease in lung cancer mortality observed in The National Lung Screening Trial Research Team study involving lung tumor screening using low-dose computed tomography (CT) may relate at least in part to low-dose X-rays activating the body's natural anticancer defenses (i.e., radiation hormesis). This possibility was apparently not recognized by the indicated research team.
Collapse
Affiliation(s)
- B R Scott
- Lovelace Respiratory Research Institute, 2425 Ridgecrest Drive SE, Albuquerque, NM 87108
| | | | | | | | | |
Collapse
|