Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 422
1.
Radiat Res ; 200(6): 556-568, 2023 Dec 01.
Article En | MEDLINE | ID: mdl-37874034

Ionizing radiation in space, radiation devices or nuclear disasters are major threats to human health and public security. Expanding countermeasures for dealing with accidental or occupational radiation exposure is crucial for the protection of radiation injuries. Circulating microRNAs (miRNAs) have emerged as promising radiation biomarkers in recent years. However, the origin, distribution and functions of radiosensitive circulating miRNAs remain unclear, which obstructs their clinical applications in the future. In this study, we found that mmu-miR-342-3p (miR-342) in mouse serum presents a stable and significant decrease after X-ray total-body irradiation (TBI). Focusing on this miRNA, we investigated the influences of circulating miR-342 on the radiation-induced injury. Through tail vein injection of Cy5-labeled synthetic miR-342, we found the exogenous miR-342-Cy5 was mainly enriched in metabolic and immune organs. Besides, the bioinformatic analysis predicted that miR-342 might involve in immune-related processes or pathways. Further, mice were tail vein injected with synthetic miR-342 mimetics (Ago-miR-342) after irradiation to upregulate the level of miR-342 in circulating blood. The results showed that the upregulation of circulating miR-342 alleviated the radiation-induced depletion of CD3+CD4+ T lymphocytes and influenced the levels of IL-2 and IL-6 in irradiated mice. Moreover, the injection of Ago-miR-342 improved the survival rates of mice with acute radiation injury. Our findings demonstrate that upregulation of circulating miR-342 alleviates the radiation-induced immune system injury, which provides us new insights into the functions of circulating miRNAs and the prospect as the targets for mitigation of radiation injuries.


Circulating MicroRNA , MicroRNAs , Radiation Injuries , Animals , Mice , Biomarkers , Circulating MicroRNA/genetics , Circulating MicroRNA/metabolism , Immune System/radiation effects , MicroRNAs/genetics , Radiation Injuries/genetics
2.
Brachytherapy ; 22(1): 21-29, 2023.
Article En | MEDLINE | ID: mdl-36437221

PURPOSE: Whether prostate brachytherapy (BT) results in opportunistic biological changes that can improve clinical outcomes is not well studied. We sought to investigate the impact of prostate BT on the immune system. MATERIALS AND METHODS: A scoping review was performed using PubMed/Scopus for papers published between 2011-2021. Search terms were "brachytherapy" AND "immune" AND "prostate". A total of 81 records were identified and 6 were selected for further review. RESULTS: 2 low-dose-rate BT papers (n=68) evaluated changes in the peripheral blood following I-125 monotherapy. Both showed significant increases in peripheral CD3+ and CD4+ T cells post-BT. One also demonstrated significant increases in Treg subsets up to 150 days post-BT. 4 high-dose-rate (HDR) studies (n=37) were identified, and all were done in combination with EBRT. The largest study (n=24) showed a single 10 Gy fraction of HDR converted 80% of "cold" tumors into an "intermediate" or "hot" state, based on a tumor inflammation signature when comparing a pre-BT biopsy to one prior to a second HDR fraction. CONCLUSION: Prostate BT can invoke an immune activating phenotype; however, changes in immunosuppressive cells are also seen. Additional data is needed to understand how to promote synergy between BT and the immune system.


Brachytherapy , Immune System , Immunity , Prostatic Neoplasms , Humans , Male , Brachytherapy/methods , Immunity/radiation effects , Iodine Radioisotopes/therapeutic use , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Radiotherapy Dosage , Immune System/radiation effects
3.
Int J Radiat Biol ; 99(3): 406-418, 2023.
Article En | MEDLINE | ID: mdl-35759247

PURPOSE: The aim of the present study was to investigate the injuries of spleen and intestinal immune system induced by 2 Gy 60Co γ ray in mice. MATERIALS AND METHODS: A total of 120 Balb/c mice were randomly divided into two groups: blank control (Ctrl) and model (IR). The IR mice were exposed to a single dose of total body irradiation (2 Gy, dose rate: 1 Gy/min) and sacrificed on 1st, 3rd, 7th, 14th and 21st day after irradiation. The indicators including general observations and body weight, the changes in peripheral hemogram, spleen index, histopathology examination and lymphocyte subsets of spleen. As well as the count and subsets of lymphocyte in gut-associated lymphoid tissue. RESULTS: Compared with the Ctrl group, the body weight, spleen index, peripheral blood cell and splenocyte amounts, intraepithelial lymphocytes number decreased significantly after exposure, accompanied by a notable decreased count of lymphocytes in Peyer's patch and mesenteric lymph nodes. Moreover, ionizing radiation also broke the balance of CD4+/CD8+ and increased the Treg proportion in spleen, which then triggered immune imbalance and immunosuppression. In general, the spleen injuries occurred on 1st day after exposure, worse on 3rd day, and were relieved on 7th day. The intestinal immune injuries were observed on 1st day, and attenuated on 3rd day. On 21st day after exposure, the spleen volume and index have returned to normal, except for the distribution of lymphocyte subpopulations. Furthermore, all indicators of gut-associated lymphoid tissue, except for mesenteric lymph nodes lymphocyte count, had returned to normal levels on 21st day. CONCLUSION: In conclusion, our data showed the injuries of spleen and intestinal immune system induced by 2 Gy 60Co γ ray whole-body irradiation. These findings may provide the bases for further radiation protection in the immunity.


Spleen , Whole-Body Irradiation , Mice , Animals , Spleen/radiation effects , Whole-Body Irradiation/adverse effects , Gamma Rays/adverse effects , Immune System/radiation effects , Body Weight
4.
Int J Clin Oncol ; 28(2): 201-208, 2023 Feb.
Article En | MEDLINE | ID: mdl-35556190

The effects of irradiation on tumor tissue and the host immune system are interrelated. The antitumor effect of irradiation is attenuated in the immunocompromised hosts. In addition, radiation alone positively and negatively influences the host immune system. The positive effects of radiation are summarized by the ability to help induce and enhance tumor-antigen-specific immune responses. The cancer-immunity cycle is a multistep framework that illustrates how the tumor-antigen-specific immune responses are induced and how the induced antigen-specific immune cells exert their functions in tumor tissues. Irradiation affects each step of this cancer-immunity cycle, primarily in a positive manner. In contrast, radiation also has negative effects on the immune system. The first is that irradiation has the possibility to kill irradiated effector immune cells. The second is that irradiation upregulates immunosuppressive molecules in the tumor microenvironment, whereas the third is that irradiation to the tumor condenses immunosuppressor cells in the tumor microenvironment. When used in conjunction with radiotherapy, immune checkpoint inhibitors can further leverage the positive effects of radiation on the immune system and compensate for the negative effects of irradiation, which supports the rationale for the combination of radiotherapy and immune checkpoint inhibitors. In this review, we summarize the preclinical evidence for the reciprocal effects of radiation exposure and the immune system, and up-front topics of the combination therapy of immune checkpoint inhibitors and radiotherapy.


Immune Checkpoint Inhibitors , Neoplasms , Humans , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Antigens, Neoplasm , Immune System/pathology , Immune System/radiation effects , Combined Modality Therapy , Tumor Microenvironment , Immunotherapy , Radiotherapy
5.
Cancer Immunol Immunother ; 71(3): 541-552, 2022 Mar.
Article En | MEDLINE | ID: mdl-34269847

PURPOSE: The influence of radiotherapy on patient immune cell subsets has been established by several groups. Following a previously published analysis of immune changes during and after curative radiotherapy for prostate cancer, this analysis focused on describing correlations of changes of immune cell subsets with radiation treatment parameters. PATIENTS AND METHODS: For 13 patients treated in a prospective trial with radiotherapy to the prostate region (primary analysis) and five patients treated with radiotherapy to prostate and pelvic nodal regions (exploratory analysis), already published immune monitoring data were correlated with clinical data as well as radiation planning parameters such as clinical target volume (CTV) and volumes receiving 20 Gy (V20) for newly contoured volumes of pelvic blood vessels and bone marrow. RESULTS: Most significant changes among immune cell subsets were observed at the end of radiotherapy. In contrast, correlations of age and CD8+ subsets (effector and memory cells) were observed early during and 3 months after radiotherapy. Ratios of T cells and T cell proliferation compared to baseline correlated with CTV. Early changes in regulatory T cells (Treg cells) and CD8+ effector T cells correlated with V20 of blood vessels and bone volumes. CONCLUSIONS: Patient age as well as radiotherapy planning parameters correlated with immune changes during radiotherapy. Larger irradiated volumes seem to correlate with early suppression of anti-cancer immunity. For immune cell analysis during normofractionated radiotherapy and correlations with treatment planning parameters, different time points should be looked at in future projects. TRIAL REGISTRATION NUMBER: NCT01376674, 20.06.2011.


Biomarkers , Immune System/radiation effects , Prostatic Neoplasms/immunology , Prostatic Neoplasms/radiotherapy , Radiotherapy Dosage , Radiotherapy Planning, Computer-Assisted , Adult , Age Factors , Humans , Immunophenotyping , Leukocyte Count , Lymphocyte Subsets/metabolism , Lymphocyte Subsets/radiation effects , Male , Middle Aged , Neoplasm Staging , Prostatic Neoplasms/pathology , Radiotherapy, Image-Guided , Young Adult
6.
Int J Mol Sci ; 22(18)2021 Sep 17.
Article En | MEDLINE | ID: mdl-34576217

Radiation therapy (RT) recruits myeloid cells, leading to an immunosuppressive microenvironment that impedes its efficacy against tumors. Combination of immunotherapy with RT is a potential approach to reversing the immunosuppressive condition and enhancing tumor control after RT. This study aimed to assess the effects of local interleukin-12 (IL-12) therapy on improving the efficacy of RT in a murine prostate cancer model. Combined treatment effectively shrunk the radioresistant tumors by inducing a T helper-1 immune response and influx of CD8+ T cells. It also delayed the radiation-induced vascular damage accompanied by increased α-smooth muscle actin-positive pericyte coverage and blood perfusion. Moreover, RT significantly reduced the IL-12-induced levels of alanine aminotransferase in blood. However, it did not further improve the IL-12-induced anti-tumor effect on distant tumors. Upregulated expression of T-cell exhaustion-associated genes was found in tumors treated with IL-12 only and combined treatment, suggesting that T-cell exhaustion is potentially correlated with tumor relapse in combined treatment. In conclusion, this study illustrated that combination of radiation and local IL-12 therapy enhanced the host immune response and promoted vascular maturation and function. Furthermore, combination treatment was associated with less systemic toxicity than IL-12 alone, providing a potential option for tumor therapy in clinical settings.


Immune System/radiation effects , Interleukin-12 Subunit p35/metabolism , Radiotherapy/methods , Actins/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Immunohistochemistry , Immunosuppressive Agents/pharmacology , Immunotherapy , Interferon-gamma/metabolism , Liver/metabolism , Liver/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth/metabolism , Neoplasm Transplantation , Pericytes/metabolism , Prostatic Neoplasms/metabolism , Tumor Microenvironment/immunology
7.
Cancer Radiother ; 25(6-7): 533-536, 2021 Oct.
Article Fr | MEDLINE | ID: mdl-34462213

The abscopal effect has been mentioned since 1953. The increase in knowledge about the immune system and the development of immunotherapies support its potential therapeutic interest. While it is accepted that radiotherapy induces an immune response, demonstrating its systemic impact is not easy. The preclinical basis is solid but its clinical validation pending. Radiotherapy rarely induces tumor reduction at a distance from the beams, probably due to its immunosuppressive effect. This is why a synergy between radiotherapy and systemic treatments targeting these immunosuppressive mechanisms was observed. Several parameters can modulate the induction of the abscopal effect. Among these, the fractionation of the dose seems to be determining with currently a pre-eminence of hypofractionated stereotaxis. On the other hand, even if the choice of more immunogenic targets (liver, lung) should be favoured, the optimal number of lesions to be irradiated remains to be defined as well as the minimum volume allowing sufficient release of tumor antigens. The impact of radiation-induced lymphopenia on radiotherapy/immunotherapy efficacy needs to be assessed more precisely, as does the effect of radiotherapy techniques on them. Finally, the choice of immunotherapy(ies) and the combination regimen with radiotherapy remain under discussion. A sequential scheme appears to provide less toxicities but the concomitant would lead to a better response. The study of these different parameters should allow us to deliver optimized radiotherapy/immunotherapy(ies) combinations to our metastatic patients in order to benefit as many people as possible from this abscopal effect.


Immunotherapy/methods , Neoplasm Metastasis/radiotherapy , Radiotherapy/methods , Antigens, Neoplasm/immunology , Combined Modality Therapy/methods , Dose Fractionation, Radiation , Humans , Immune System/radiation effects , Immunosuppression Therapy , Lymphopenia/immunology , Neoplasm Metastasis/immunology , Neoplasms/immunology , Neoplasms/radiotherapy , Radiotherapy/adverse effects , Treatment Outcome
8.
Int J Mol Sci ; 22(9)2021 Apr 27.
Article En | MEDLINE | ID: mdl-33925430

Although ionizing radiation (radiation) is commonly used for medical diagnosis and cancer treatment, radiation-induced damages cannot be avoided. Such damages can be classified into direct and indirect damages, caused by the direct absorption of radiation energy into DNA and by free radicals, such as hydroxyl radicals (•OH), generated in the process of water radiolysis. More specifically, radiation damage concerns not only direct damages to DNA, but also secondary damages to non-DNA targets, because low-dose radiation damage is mainly caused by these indirect effects. Molecular hydrogen (H2) has the potential to be a radioprotective agent because it can selectively scavenge •OH, a reactive oxygen species with strong oxidizing power. Animal experiments and clinical trials have reported that H2 exhibits a highly safe radioprotective effect. This paper reviews previously reported radioprotective effects of H2 and discusses the mechanisms of H2, not only as an antioxidant, but also in intracellular responses including anti-inflammation, anti-apoptosis, and the regulation of gene expression. In doing so, we demonstrate the prospects of H2 as a novel and clinically applicable radioprotective agent.


Hydrogen/pharmacology , Neoplasms/therapy , Radiation Injuries/prevention & control , Radiation-Protective Agents/pharmacology , Animals , Antioxidants/pharmacology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/prevention & control , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Humans , Hydrogen/therapeutic use , Immune System/drug effects , Immune System/radiation effects , Male , Quality of Life , Radiation-Protective Agents/therapeutic use , Skin/drug effects , Skin/radiation effects , Spermatozoa/drug effects , Spermatozoa/radiation effects
9.
Clin Transl Oncol ; 23(9): 1827-1837, 2021 Sep.
Article En | MEDLINE | ID: mdl-33774805

BACKGROUND: Poly-(ADP-Ribose)-Polymerase inhibitors (PARPi) were reported as radiosensitizers in non-small cell lung cancer (NSCLC) with wide-type epidermal growth factor receptor (EGFR), but the effects of radiation combined with PARPi were not investigated in EGFR-mutated NSCLC. Moreover, the underlying mechanisms were not well examined. This study aimed to study the efficacy of radiation combined with niraparib in EGFR-mutated NSCLC and explore their influence on the immune system. METHODS: Clone formation and apoptosis assay were conducted to explore the effects of niraparib and radiation. Immunofluorescence was conducted to detect the double-strand DNA breaks. Real-time PCR and immunoblotting were employed to evaluate the activation of STING/TBK1/TRF3 pathway and the expression levels of interferon ß, CCL5 and CXCL10. Immunocompetent mice model bearing with subcutaneous Lewis lung cancer was established to confirm the results in vivo. RESULTS: Niraparib and radiation were synergistic to inhibit tumor both in vitro and in vivo. Radiation plus niraparib could activate anti-tumor immunity, which appeared as increased CD8+ T lymphocytes and activated STING/TBK1/IRF3 pathway. CONCLUSION: PARPi not only as a radiosensitizer inhibited EGFR-mutated NSCLC tumor growth, but also cooperated with radiation to promote anti-tumor immune responses.


Carcinoma, Non-Small-Cell Lung/therapy , Chemoradiotherapy/methods , Genes, erbB-1 , Indazoles/pharmacology , Lung Neoplasms/therapy , Mutation , Piperidines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , CD8-Positive T-Lymphocytes , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Chemokine CCL5/metabolism , Chemokine CXCL10/metabolism , DNA Breaks, Double-Stranded , Female , Fluorescent Antibody Technique , Humans , Immune System/drug effects , Immune System/radiation effects , Immunocompetence , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lymphocytes, Tumor-Infiltrating , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Radiation Tolerance/drug effects , Real-Time Polymerase Chain Reaction , TATA Box Binding Protein-Like Proteins/metabolism , Tumor Stem Cell Assay
10.
J Appl Microbiol ; 130(1): 14-24, 2021 Jan.
Article En | MEDLINE | ID: mdl-32692438

The gut microbiome is well recognized to have a pivotal role in regulation of the health and behaviour of the host, affecting digestion, metabolism, immunity, and has been linked to changes in bones, muscles and the brain, to name a few. However, the impact of microgravity environment on gut bacteria is not well understood. In space environments, astronauts face several health issues including stress, high iron diet, radiation and being in a closed system during extended space missions. Herein, we discuss the role of gut bacteria in the space environment, in relation to factors such as microgravity, radiation and diet. Gut bacteria may exact their effects by synthesis of molecules, their absorption, and through physiological effects on the host. Moreover we deliberate the role of these challenges in the dysbiosis of the human microbiota and possible dysregulation of the immune system.


Extraterrestrial Environment , Gastrointestinal Microbiome/physiology , Diet/adverse effects , Dysbiosis/physiopathology , Humans , Immune System/physiology , Immune System/radiation effects , Radiation , Space Flight , Stress, Physiological/physiology , Stress, Physiological/radiation effects , Weightlessness/adverse effects
11.
Radiat Res ; 194(4): 411-430, 2020 10 02.
Article En | MEDLINE | ID: mdl-32936898

In the event of a major accidental or intentional radiation exposure incident, the affected population could suffer from total- or partial-body exposures to ionizing radiation with acute exposure to organs that would produce life-threatening injury. Therefore, it is necessary to identify markers capable of predicting organ-specific damage so that appropriate directed or encompassing therapies can be applied. In the current work, gene expression changes in response to total-body irradiation (TBI) were identified in heart, lungs and liver tissue of Göttingen minipigs. Animals received 1.7, 1.9, 2.1 or 2.3 Gy TBI and were followed for 45 days. Organ samples were collected at the end of day 45 or sooner if the animal displayed morbidity necessitating euthanasia. Our findings indicate that different organs respond to TBI in a very specific and distinct manner. We also found that the liver was the most affected organ in terms of gene expression changes, and that lipid metabolic pathways were the most deregulated in the liver samples of non-survivors (survival time <45 days). We identified organ-specific gene expression signatures that accurately differentiated non-survivors from survivors and control animals, irrespective of dose and time postirradiation. At what point did these radiation-induced injury markers manifest and how this information could be used for applying intervention therapies are under investigation.


Gene Expression Profiling , Heart/radiation effects , Liver/radiation effects , Lung/radiation effects , Radiation Injuries, Experimental/genetics , Whole-Body Irradiation/adverse effects , Animals , Apelin/physiology , Cobalt Radioisotopes , Computer Systems , Dose-Response Relationship, Radiation , Endothelium, Vascular/embryology , Endothelium, Vascular/radiation effects , Gamma Rays/adverse effects , Immune System/radiation effects , Kaplan-Meier Estimate , Lipid Metabolism/radiation effects , Liver/metabolism , Lung/immunology , Lung/metabolism , Male , Myocardium/metabolism , Oligonucleotide Array Sequence Analysis , Organ Specificity , Phantoms, Imaging , Radiation Injuries, Experimental/etiology , Signal Transduction/radiation effects , Swine , Swine, Miniature
13.
Strahlenther Onkol ; 196(11): 1018-1033, 2020 Nov.
Article En | MEDLINE | ID: mdl-32519025

BACKGROUND: In this exploratory study, the impact of local irradiation on systemic changes in stress and immune parameters was investigated in eight patients treated with intensity-modulated radiation therapy (IMRT) or stereotactic ablative body radiotherapy (SABR) for prostate adenocarcinoma to gain deeper insights into how radiotherapy (RT) modulates the immune system. PATIENTS AND METHODS: RT-qPCR, flow cytometry, metabolomics, and antibody arrays were used to monitor a panel of stress- and immune-related parameters before RT, after the first fraction (SABR) or the first week of treatment (IMRT), after the last fraction, and 3 weeks later in the blood of IMRT (N = 4) or SABR (N = 4) patients. Effect size analysis was used for comparison of results at different timepoints. RESULTS: Several parameters were found to be differentially modulated in IMRT and SABR patients: the expression of TGFB1, IL1B, and CCL3 genes; the expression of HLA-DR on circulating monocytes; the abundance and ratio of phosphatidylcholine and lysophosphatidylcholine metabolites in plasma. More immune modulators in plasma were modulated during IMRT than SABR, with only two common proteins, namely GDF-15 and Tim­3. CONCLUSION: Locally delivered RT induces systemic modulation of the immune system in prostate adenocarcinoma patients. IMRT and SABR appear to specifically affect distinct immune components.


Adenocarcinoma/radiotherapy , Adenocarcinoma/surgery , Immune System/radiation effects , Metabolome/radiation effects , Neoplasm Proteins/blood , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/surgery , Proteome/radiation effects , Radiosurgery/methods , Radiotherapy, Intensity-Modulated/methods , Stress, Physiological/radiation effects , Adenocarcinoma/immunology , Adenocarcinoma/physiopathology , Aged , Aged, 80 and over , Biomarkers , Cytokines/blood , Gene Expression Regulation, Neoplastic/radiation effects , HLA Antigens/blood , Humans , Inflammation Mediators/blood , Lysophosphatidylcholines/blood , Male , Middle Aged , Monocytes/immunology , Phosphatidylcholines/blood , Prostatic Neoplasms/immunology , Prostatic Neoplasms/physiopathology
14.
Biomed Res Int ; 2020: 2641324, 2020.
Article En | MEDLINE | ID: mdl-32566675

During spaceflight, the homeostasis of the living body is threatened with cosmic environment including microgravity and irradiation. Traditional Chinese medicine could ameliorate the internal imbalance during spaceflight, but its mechanism is still unclear. In this article, we compared the difference of neuroendocrine-immune balance between simulated microgravity (S) and simulated microgravity and irradiation (SAI) environment. We also observed the antagonistic effect of SAI using a traditional Chinese medicine formula (TCMF). Wistar rats were, respectively, exposed under S using tail suspending and SAI using tail suspending and 60Co-gama irradiation exposure. The SAI rats were intervened with TCMF. The changes of hypothalamic-pituitary-adrenal (HPA) axis, splenic T-cell, celiac macrophages, and related cytokines were observed after 21 days. Compared with the normal group, the hyperfunction of HPA axis and celiac macrophages, as well as the hypofunction of splenic T-cells, was observed in both the S and SAI group. Compared with the S group, the levels of plasmatic corticotropin-releasing hormone (CRH), macrophage activity, and serous interleukin-6 (IL-6) in the SAI group were significantly reduced. The dysfunctional targets were mostly reversed in the TCMF group. Both S and SAI could lead to NEI imbalance. Irradiation could aggravate the negative feedback inhibition of HPA axis and macrophages caused by S. TCMF could ameliorate the NEI dysfunction caused by SAI.


Drugs, Chinese Herbal/pharmacology , Immune System , Neurosecretory Systems , Weightlessness , Animals , Corticosterone/metabolism , Corticotropin-Releasing Hormone/metabolism , Cytokines/metabolism , Gamma Rays , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/radiation effects , Immune System/drug effects , Immune System/radiation effects , Macrophages/drug effects , Macrophages/radiation effects , Male , Medicine, Chinese Traditional , Models, Biological , Neurosecretory Systems/drug effects , Neurosecretory Systems/radiation effects , Rats , Rats, Wistar , T-Lymphocytes/drug effects , T-Lymphocytes/radiation effects
15.
Oncology ; 98(4): 202-215, 2020.
Article En | MEDLINE | ID: mdl-32079015

BACKGROUND: Malignant melanoma represents the deadliest form of skin cancer with a high tendency to metastasize during the early course of the disease. Radiation therapy has long played a key role in the management of both local and metastatic melanoma. Although local radiation therapy exerts antitumor effects by damaging the cellular DNA, it also induces an important out-of-field (distant) effect known as the "abscopal effect" in nonirradiated sites. Radiation therapy-induced abscopal effects are believed to be mediated by activation and stimulation of the immune system. OBJECTIVE: To provide a detailed overview of the current state of knowledge and clinical experience of radiation therapy-induced abscopal effects in patients with malignant melanoma. METHODS: Using electronic databases such as MEDLINE via PubMed and Google Scholar, a systematic literature review was performed to find published clinical evidence for radiation therapy-induced abscopal effects in patients with malignant melanoma. The clinical data on radiation therapy-induced abscopal effects were reviewed and the outcomes summarized. RESULTS: Clinical evidence of patients with malignant melanoma was gathered using databases from MEDLINE and those findings were summarized. Although the precise mechanism of the abscopal effect of radiation therapy is still not completely understood, evidence suggests that tumor cell destruction by radiation releases tumor antigens that stimulate the immune system of the host to activate the body's immune effector cells systemically and produce distant non-target antitumor effects. This forms a basis for using the radiation therapy with immunotherapy to augment the abscopal response rates. CONCLUSIONS: Current clinical evidence suggests that there is a large potential to enhance the abscopal effect when radiation therapy is combined with immunotherapeutic agents for the treatment of malignant melanoma. Ongoing and planned clinical trials may provide us with a more in-depth understanding of how this combination therapy can be optimally utilized clinically to achieve improved survival outcomes among patients with malignant melanoma.


Bystander Effect , Immune System/radiation effects , Melanoma/radiotherapy , Humans
16.
Front Immunol ; 11: 614960, 2020.
Article En | MEDLINE | ID: mdl-33552079

Dim light at night (dLAN) has become a pervasive part of the modern world, and growing evidence shows its association with increased health risks. Though this link is attributed to a disturbed circadian clock, the underlying mechanisms that can explain how circadian disruption from dLAN causes negative health effects remain unclear. Here, we exposed rats to a light-dark cycle (12:12 h) with low-intensity light at night (~2 lx) for 2 and 5 weeks and explored the steady-state pattern of circulating immune cells and renal immune-related markers, which are well controlled by the circadian clock. After 5 weeks, dLAN impaired the daily variation in several types of white blood cells, especially monocytes and T cells. Two-week dLAN caused a reduction in blood monocytes and altered gene expression of macrophage marker Cd68 and monocyte-attracting chemokine Ccl2 in the kidney. Interestingly, dLAN decreased renal 3-nitrotyrosine levels and resulted in up-regulation of the main endogenous antioxidant pathways, indicating a disturbance in the renal redox balance and an activation of compensatory mechanisms. These effects paralleled the altered renal expression of the molecular clock components and increased plasma corticosterone levels. Together, our results show that chronic exposure to dLAN weakened the circadian control of daily variation of circulating immune cells and disturbed renal immune and redox homeostasis. Consequences of this dLAN-disturbed immune balance on the ability of the immune system to cope with other challenges should by clarified in further studies.


Circadian Rhythm/immunology , Immune System/radiation effects , Kidney/immunology , Light/adverse effects , Photoperiod , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/biosynthesis , Antigens, Differentiation, Myelomonocytic/genetics , CLOCK Proteins/biosynthesis , CLOCK Proteins/genetics , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Chemokines/biosynthesis , Chemokines/genetics , Corticosterone/blood , Gene Expression Regulation/radiation effects , Homeostasis/radiation effects , Immunophenotyping , Kidney/metabolism , Kidney Cortex/enzymology , Leukocyte Count , Male , Melatonin/blood , Oxidation-Reduction , Rats , Rats, Wistar , Respiratory Burst , Superoxide Dismutase/analysis
17.
Brachytherapy ; 19(1): 51-59, 2020.
Article En | MEDLINE | ID: mdl-31690516

PURPOSE: We hypothesized that radiation doses delivered with high-dose-rate (HDR) and pulsed-dose-rate (PDR) brachytherapy in patients with cervical cancer could trigger immune stimulation by modulating immune cells in the tumor microenvironment. The objective was to determine CD68, CD163, and PD-L1 expression in biopsies from patients with cervical cancer and to compare the effects of HDR vs. PDR brachytherapy on the expression of these proteins. METHODS AND MATERIALS: Nineteen consecutive women (mean age, 55.9 years) with histologically proven cervical cancer scheduled for brachytherapy after standard external beam irradiation therapy combined with platinum-based chemotherapy were included in a prospective study. Core tissue biopsies were obtained before radiochemotherapy (biopsy #1), after completion of radiochemotherapy and before brachytherapy (biopsy #2), and 2 weeks after completion of brachytherapy (biopsy #3). HDR or PDR brachytherapy was delivered according to availability of equipment. CD68, CD163, and PD-L1 immunohistochemical expression was estimated using a quantitative method. Conditional logistic regression models were used to assess the relationship between gene expression and time of biopsy for each brachytherapy technique. RESULTS: In relation to CD68 and CD163, statistically significant relationships between gene expression and biopsy tissue samples were not found in any of the brachytherapy techniques, although there was trend toward downexpression of CD68 and CD163 in biopsies #2 and #3 in the HDR brachytherapy cohort only. There was a significant increase in PD-L1 expression in biopsy #3 also in the HDR cohort as compared with the PDR cohort. CONCLUSIONS: Decreased CD68 and CD163 expression did not reach statistical significance, but this trend may have clinical translational potential. Overexpression of PD-L1 in tissue biopsies taken at 14 days in the HDR brachytherapy cohort may tentatively suggest that this time period would be an adequate interval for blockade of the PD-1/PD-L1 axis.


Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , B7-H1 Antigen/metabolism , Brachytherapy/methods , Receptors, Cell Surface/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/radiotherapy , Adult , Aged , Chemoradiotherapy , Female , Humans , Immune System/radiation effects , Middle Aged , Prospective Studies , Radiotherapy Dosage , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Uterine Cervical Neoplasms/drug therapy
18.
Immunotherapy ; 11(17): 1445-1461, 2019 12.
Article En | MEDLINE | ID: mdl-31826745

The abscopal effect describes the ability of locally administered radiotherapy to induce systemic antitumor effects. Over the past 40 years, reports on the abscopal effect following conventional radiation have been relatively rare, especially in less immunogenic tumors such as lung cancer. However, with the continued development and use of immunotherapy, reports on the abscopal effect have become increasingly frequent during the last decade. Here, we present three illustrative case reports from our own institution and previous published cases of the abscopal effect in patients with non-small cell lung cancer, treated with immune checkpoint inhibitors and radiotherapy. We also present a concise review of the clinical and experimental literature on the abscopal effect in non-small cell lung cancer.


Immune System/radiation effects , Lung Neoplasms/radiotherapy , Aged , Humans , Immunotherapy , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Middle Aged , Radiotherapy Dosage , Tomography, X-Ray Computed
19.
Mol Ecol ; 28(20): 4620-4635, 2019 10.
Article En | MEDLINE | ID: mdl-31498518

Wildlife inhabiting environments contaminated by radionuclides face putative detrimental effects of exposure to ionizing radiation, with biomarkers such as an increase in DNA damage and/or oxidative stress commonly associated with radiation exposure. To examine the effects of exposure to radiation on gene expression in wildlife, we conducted a de novo RNA sequencing study of liver and spleen tissues from a rodent, the bank vole Myodes glareolus. Bank voles were collected from the Chernobyl Exclusion Zone (CEZ), where animals were exposed to elevated levels of radionuclides, and from uncontaminated areas near Kyiv, Ukraine. Counter to expectations, we did not observe a strong DNA damage response in animals exposed to radionuclides, although some signs of oxidative stress were identified. Rather, exposure to environmental radionuclides was associated with upregulation of genes involved in lipid metabolism and fatty acid oxidation in the livers - an apparent shift in energy metabolism. Moreover, using stable isotope analysis, we identified that fur from bank voles inhabiting the CEZ had enriched isotope values of nitrogen: such an increase is consistent with increased fatty acid metabolism, but also could arise from a difference in diet or habitat between the CEZ and elsewhere. In livers and spleens, voles inhabiting the CEZ were characterized by immunosuppression, such as impaired antigen processing, and activation of leucocytes involved in inflammatory responses. In conclusion, exposure to low dose environmental radiation impacts pathways associated with immunity and lipid metabolism, potentially as a stress-induced coping mechanism.


Chernobyl Nuclear Accident , Lipid Metabolism/physiology , Liver/pathology , Radiation Exposure/adverse effects , Spleen/pathology , Animals , Arvicolinae , DNA Damage/radiation effects , DNA Repair/radiation effects , Fatty Acids/metabolism , Immune System/radiation effects , Lipid Metabolism/genetics , Liver/immunology , Mutagens/adverse effects , Oxidation-Reduction/radiation effects , Oxidative Stress/radiation effects , Radiation, Ionizing , Radioisotopes/adverse effects , Spleen/immunology , Ukraine
20.
Ann Palliat Med ; 8(3): 326-336, 2019 Jul.
Article En | MEDLINE | ID: mdl-31370663

Radiation therapy is commonly used in the metastatic setting to palliate pain, neurological deficits, bleeding and other complications of metastatic disease, allowing patients to live longer and have better quality of life. Despite the effective use of radiation and other palliative treatment modalities, many patients continue to experience poorly controlled pain and other serious sequelae of their disease, underscoring the need for additional research in this area. In this review we highlight recent developments impacting the fields of palliative care and radiation oncology and describe opportunities for research and innovation including studies of tumor microenvironment, identification of effective biomarkers of tumor response and combinatorial treatments with new systemic agents. It is our hope that progress in these fields will improve the lives of patients living with advanced malignancies.


Antineoplastic Agents, Immunological/therapeutic use , Cancer Pain/radiotherapy , Neoplasms/radiotherapy , Palliative Care/methods , Translational Research, Biomedical/organization & administration , Animals , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor , Bone Neoplasms/radiotherapy , Bone Neoplasms/secondary , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Cancer-Associated Fibroblasts/radiation effects , Combined Modality Therapy , Humans , Immune System/radiation effects , Life Expectancy , Neoplasm Metastasis , Neoplasms/pathology , Neoplasms/therapy , Quality of Life , Radiation Oncology , Spinal Neoplasms/radiotherapy , Spinal Neoplasms/secondary , Tumor Microenvironment/radiation effects
...