Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 702
Filtrar
1.
J Transl Med ; 22(1): 519, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38816831

RESUMEN

BACKGROUND: Telomerase, by safeguarding damaged telomeres and bolstering DNA damage repair, has the capacity to heighten the radioresistance of tumour cells. Thus, in turn, can compromise the efficacy of radiotherapy (RT) and radioimmunotherapy. Our previous studies have revealed that the highly selective telomerase inhibitor, BIBR1532, possesses the potential to enhance the radiosensitivity of Non-small cell lung cancer (NSCLC). In this study, we delve further into the impact of BIBR1532 on the immune activation induced by RT and elucidate the underlying mechanisms. METHODS: Biological information analyses, immunofluorescence assays, western blot assays, flow cytometry analysis were conducted to elucidate the functions of the combination of BIBR1532 with radiotherapy in NSCLC. Intracellular levels of lipid peroxides, glutathione, malondialdehyde, and Fe2+ were measured as indicators of ferroptosis status. Both in vitro and in vivo studies were conducted to examine the antitumor effects. RESULTS: Our findings indicate that the confluence of BIBR1532 with RT significantly augments the activation of the cGAS-STING pathway in both in vivo and in vitro settings, thereby fostering an effective anti-tumoral immune response. The effects can be ascribed to two key processes. Firstly, ionizing radiation, in precipitating DNA double-strand breaks (DSBs), prompts the release of tumour-derived double-stranded DNA (dsDNA) into the cytoplasm. Subsequently, BIBR1532 amplifies the activation of antigen-presenting cells by dsDNA post-RT and instigates the cGAS-STING pathway. Secondly, BIBR1532 enhances the ferroptosis response in NSCLC following RT, thereby promoting unrestrained lipid peroxidation and elevated levels of reactive oxygen species (ROS) within tumour cells. This ultimately leads to mitochondrial stress and the release of endogenous mitochondrial DNA (mtDNA) into the cytoplasm, thus facilitating the activation of the STING pathway and the induction of a type I interferon (IFN)-linked adaptive immune response. CONCLUSION: This study underscores the potential of BIBR1532 as an efficacious and safe radiosensitizer and radioimmunotherapy synergist, providing robust preclinical research evidence for the treatment of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Ferroptosis , Neoplasias Pulmonares , Proteínas de la Membrana , Nucleotidiltransferasas , Transducción de Señal , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/inmunología , Proteínas de la Membrana/metabolismo , Transducción de Señal/efectos de la radiación , Nucleotidiltransferasas/metabolismo , Línea Celular Tumoral , Animales , Inmunidad/efectos de la radiación , Ratones Desnudos , Ratones
2.
Brachytherapy ; 22(1): 21-29, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36437221

RESUMEN

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.


Asunto(s)
Braquiterapia , Sistema Inmunológico , Inmunidad , Neoplasias de la Próstata , Humanos , Masculino , Braquiterapia/métodos , Inmunidad/efectos de la radiación , Radioisótopos de Yodo/uso terapéutico , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/radioterapia , Dosificación Radioterapéutica , Sistema Inmunológico/efectos de la radiación
3.
Cancer Sci ; 113(4): 1352-1361, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35133062

RESUMEN

Radiotherapy (RT) combined with immune checkpoint inhibitors has recently produced outstanding results and is expected to be adaptable for various cancers. However, the precise molecular mechanism by which immune reactions are induced by fractionated RT is still controversial. We aimed to investigate the mechanism of the immune response regarding multifractionated, long-term radiation, which is most often combined with immunotherapy. Two human esophageal cancer cell lines, KYSE-450 and OE-21, were irradiated by fractionated irradiation (FIR) daily at a dose of 3 Gy in 5 d/wk for 2 weeks. Western blot analysis and RNA sequencing identified type I interferon (IFN) and the stimulator of IFN genes (STING) pathway as candidates that regulate immune response by FIR. We inhibited STING, IFNAR1, STAT1, and IFN regulatory factor 1 (IRF1) and investigated the effects on the immune response in cancer cells and the invasion of surrounding immune cells. We herein revealed type I IFN-dependent immune reactions and the positive feedback of STING, IRF1, and phosphorylated STAT1 induced by FIR. Knocking out STING, IFNAR1, STAT1, and IRF1 resulted in a poorer immunological response than that in WT cells. The STING-KO KYSE-450 cell line showed significantly less invasion of PBMCs than the WT cell line under FIR. In the analysis of STING-KO cells and migrated PBMCs, we confirmed the occurrence of STING-dependent immune activation under FIR. In conclusion, we identified that the STING-IFNAR1-STAT1-IRF1 axis regulates immune reactions in cancer cells triggered by FIR and that the STING pathway also contributes to immune cell invasion of cancer cells.


Asunto(s)
Neoplasias Esofágicas , Inmunidad , Factor 1 Regulador del Interferón , Factor de Transcripción STAT1 , Línea Celular/efectos de la radiación , Neoplasias Esofágicas/genética , Humanos , Inmunidad/efectos de la radiación , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Factor 1 Regulador del Interferón/efectos de la radiación , Interferón Tipo I , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/efectos de la radiación , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Receptor de Interferón alfa y beta/efectos de la radiación , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/efectos de la radiación
4.
BMC Cancer ; 22(1): 88, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35062905

RESUMEN

BACKGROUND: Radiotherapy is a highly effective treatment for cervical cancer. Recent studies focused on the radiotherapy induced anti-tumor immunity. Whether tumor-derived extracellular vesicles (EVs) play roles in radiotherapy induced tumor associated macrophage (TAM) polarization remains unclear. MATERIALS AND METHODS: This study analysed the phenotype of macrophages in cancer tissue and peripheral blood of cervical cancer patients using flow cytometry analysis. The role of EVs from plasma of post-irradiated patients on M2-like transformed macrophages was assessed. The M1- and M2-like macrophages were assessed by expression of cell surface markers (CCR7, CD163) and intracellular cytokines (IL-10, TNFα and iNOS). The capacity of phagocytosis was assessed by PD-1 expression and phagocytosis of pHrodo Red E. coli bioparticles. RESULTS: Our results demonstrated that radiotherapy of cervical cancer induced an increase in the number of TAMs and a change in their subtype from the M2-like to the M1-like phenotype (increased expression of CCR7 and decreased expression of CD163). The EVs from plasma of post-irradiated patients facilitated the M2-like to the M1-like phenotype transition (increased expression of CCR7, TNFα and iNOS, and decreased expression of CD163 and IL-10) and increased capacity of phagocytosis (decreased PD-1 expression and increased phagocytosis of pHrodo Red E. coli bioparticles). CONCLUSIONS: Our data demonstrated that irradiation in cervical cancer patients facilitated a proinflammatory macrophage phenotype which could eventually able to mediate anti-tumor immune responses. Our findings highlight the importance of EV in the crosstalk of tumor cells and TAM upon irradiation, which potentially leading to an increased inflammatory response to cancer lesions.


Asunto(s)
Anticuerpos Antineoplásicos/efectos de la radiación , Vesículas Extracelulares/efectos de la radiación , Inmunidad/efectos de la radiación , Macrófagos Asociados a Tumores/efectos de la radiación , Neoplasias del Cuello Uterino/radioterapia , Adulto , Braquiterapia , Citocinas/efectos de la radiación , Femenino , Humanos , Persona de Mediana Edad , Fenotipo , Neoplasias del Cuello Uterino/inmunología
5.
Front Immunol ; 12: 803247, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34970277

RESUMEN

Purpose: Considering the limited data, we aimed to identify the greatest immune activation irradiated site of common metastases and response to immune checkpoint inhibitors simultaneously in non-small cell lung cancer (NSCLC). Methods: A total of 136 patients with advanced NSCLC who had received radiation to a primary or metastatic solid tumor were enrolled. We recorded blood cell counts in three time periods, before, during, and after radiotherapy (RT), and derived some blood index ratios including monocyte-to-lymphocyte ratio (MLR), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and systemic immune-inflammation index (SII). The delta-IBs were calculated as medio-IBs ÷ pre-IBs - 1. We analyzed the changes before and during RT using Spearman rank correlation test, Kruskal-Wallis rank sum test, and logistic regression analyzing their correlation with efficacy. Results: The medians of delta-MLR and delta-PLR were both the lowest while the median of delta-L was the highest in brain. Therapeutic effect evaluation showed that the objective response rate (ORR) of 48.65% (18/37) in the brain irradiation group was the highest, compared with 17.07% (7/41) in bone and 41.94% (13/31) in lung. Conclusions: In this study, results suggested that irradiation to brain has the best immune activation effect and patient outcome compared with other organs in NSCLC, and when the earlier-line ICIs were combined with RT, a better patient outcome was reached. Prospective studies are also necessary to provide more convincing evidence and standards for clinical irradiation metastases selection.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Inmunidad , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Adulto , Anciano , Biomarcadores de Tumor , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Terapia Combinada , Susceptibilidad a Enfermedades , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunidad/efectos de los fármacos , Inmunidad/efectos de la radiación , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Neoplasias Pulmonares/diagnóstico , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Metástasis de la Neoplasia , Estadificación de Neoplasias , Radioterapia/efectos adversos , Radioterapia/métodos , Resultado del Tratamiento
6.
Biochem Biophys Res Commun ; 585: 55-60, 2021 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-34784552

RESUMEN

Radiotherapy (RT) is an effective treatment option for cancer; however, its efficacy remains less than optimal in locally advanced cancer. Immune checkpoint inhibitor-based therapy, including the administration of anti-PD-L1 antibodies, is a promising approach that works synergistically with RT. Proton beam therapy and carbon-ion therapy are common options for patients with cancer. Proton and carbon ions are reported to induce an immune reaction in cancer cells; however, the underlying mechanisms remain unclear. Here, we aimed to compare the immune responses after irradiation (IR) with X-ray, protons, and carbon ions in an oesophageal cancer cell line and the underlying mechanisms. An oesophageal cancer cell line, KYSE450, was irradiated with 1 fraction/15 GyE (Gy equivalent) of X-ray, proton, or carbon-ion beams, and then, the cells were harvested for RNA sequencing and gene enrichment analysis. We also knocked out STING and STAT1 in the quest for mechanistic insights. RNA sequencing data revealed that gene expression signatures and biological processes were different in KYSE450 irradiated with X-ray, proton, and carbon-ion beams 6-24 h after IR. However, after 3 days, a common gene expression signature was detected, associated with biological pathways involved in innate immune responses. Gene knock-out experiments revealed that the STING-STAT1 axis underlies the immune reactions after IR. X-Ray, proton, and carbon-ion IRs induced similar immune responses, regulated by the STING-STAT1 axis.


Asunto(s)
Carbono , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Inmunidad/efectos de la radiación , Protones , Transcriptoma/efectos de la radiación , Rayos X , Línea Celular Tumoral , Neoplasias Esofágicas/inmunología , Neoplasias Esofágicas/patología , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/inmunología , Ontología de Genes , Humanos , Inmunidad/genética , Iones , RNA-Seq/métodos , Radiación/clasificación , Transducción de Señal/genética , Transducción de Señal/inmunología , Transducción de Señal/efectos de la radiación , Transcriptoma/inmunología
7.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34768958

RESUMEN

The development of new laser-driven electron linear accelerators, providing unique ultrashort pulsed electron beams (UPEBs) with low repetition rates, opens new opportunities for radiotherapy and new fronts for radiobiological research in general. Considering the growing interest in the application of UPEBs in radiation biology and medicine, the aim of this study was to reveal the changes in immune system in response to low-energy laser-driven UPEB whole-body irradiation in rodents. Forty male albino Wistar rats were exposed to laser-driven UPEB irradiation, after which different immunological parameters were studied on the 1st, 3rd, 7th, 14th, and 28th day after irradiation. According to the results, this type of irradiation induces alterations in the rat immune system, particularly by increasing the production of pro- and anti-inflammatory cytokines and elevating the DNA damage rate. Moreover, such an immune response reaches its maximal levels on the third day after laser-driven UPEB whole-body irradiation, showing partial recovery on subsequent days with a total recovery on the 28th day. The results of this study provide valuable insight into the effect of laser-driven UPEB whole-body irradiation on the immune system of the animals and support further animal experiments on the role of this novel type of irradiation.


Asunto(s)
Electrones/efectos adversos , Inmunidad/efectos de la radiación , Irradiación Corporal Total/efectos adversos , Animales , Médula Ósea/inmunología , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Citocinas/biosíntesis , Daño del ADN , Reparación del ADN/efectos de la radiación , Rayos Láser/efectos adversos , Leucocitos/inmunología , Leucocitos/patología , Leucocitos/efectos de la radiación , Masculino , Aceleradores de Partículas , Radiobiología , Ratas , Ratas Wistar
8.
Int J Mol Sci ; 22(19)2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34639014

RESUMEN

The current state of cancer treatment is still far from being satisfactory considering the strong impairment of patients' quality of life and the high lethality of malignant diseases. Therefore, it is critical for innovative approaches to be tested in the near future. In view of the crucial role that is played by tumor immunity, the present review provides essential information on the immune-mediated effects potentially generated by the interplay between ionizing radiation and cytotoxic antitumor agents when interacting with target malignant cells. Therefore, the radiation-dependent abscopal effect (i.e., a biological effect of ionizing radiation that occurs outside the irradiated field), the influence of cancer chemotherapy on the antigenic pattern of target neoplastic cells, and the immunogenic cell death (ICD) caused by anticancer agents are the main topics of this presentation. It is widely accepted that tumor immunity plays a fundamental role in generating an abscopal effect and that anticancer drugs can profoundly influence not only the host immune responses, but also the immunogenic pattern of malignant cells. Remarkably, several anticancer drugs impact both the abscopal effect and ICD. In addition, certain classes of anticancer agents are able to amplify already expressed tumor-associated antigens (TAA). More importantly, other drugs, especially triazenes, induce the appearance of new tumor neoantigens (TNA), a phenomenon that we termed drug-induced xenogenization (DIX). The adoption of the abscopal effect is proposed as a potential therapeutic modality when properly applied concomitantly with drug-induced increase in tumor cell immunogenicity and ICD. Although little to no preclinical or clinical studies are presently available on this subject, we discuss this issue in terms of potential mechanisms and therapeutic benefits. Upcoming investigations are aimed at evaluating how chemical anticancer drugs, radiation, and immunotherapies are interacting and cooperate in evoking the abscopal effect, tumor xenogenization and ICD, paving the way for new and possibly successful approaches in cancer therapy.


Asunto(s)
Antineoplásicos/efectos adversos , Inmunidad/efectos de los fármacos , Inmunidad/efectos de la radiación , Neoplasias/complicaciones , Neoplasias/inmunología , Radiación Ionizante , Radioterapia/efectos adversos , Animales , Antineoplásicos/uso terapéutico , Biomarcadores , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/metabolismo , Humanos , Modelos Animales , Neoplasias/terapia , Traumatismos por Radiación/etiología , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/patología , Radioterapia/métodos
9.
PLoS One ; 16(9): e0257097, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34506561

RESUMEN

Ceratitis capitata (medfly) is one of the most devastating crop pests worldwide. The Sterile Insect Technique (SIT) is a control method that is based on the mass rearing of males, their sterilization, and release in the field. However, the effectiveness of the technique depends on the quality of the released males and their fitness. We previously isolated and selected a probiotic bacteria (Enterobacter sp.), from wild-caught medflies, according to criteria that improved biological quality traits of reared medfly males.We firstly evaluated the impact of the irradiation on the expression of different immune and stress genes in the medfly sterile males. Expression was measured at differents time points ranging from 0 to 168 h after irradiation to capture the response of genes with distinct temporal expression patterns. Then, we supplemented the larval diet with previously isolated Enterobacter sp.strain, live and autoclaved at various concentrations to see whether the probiotic treatments affect, through their protective role, the gene expression level, and quality traits. The irradiation had significant effect on the genes attacin, cecropin, PGPR-LC, hsp23, and hsp70 level expression. The expression of attacin and PGPR-LC was up-regulated while that of cecropin was down-regulated. Hsp genes showed decreased levels between 0 and 18 h to peak at 72 h. However, the supplementation of the probiotic strain, either live or autoclaved, was statistically significant only for attacingene. However, significant interaction time x probiotic was noticed for attacin, cecropin, hsp23 and hsp70. The probiotic treatments also improved the quality control parameters like pupal weight. From this work we can conclude that a consortium of parabiotics (autoclaved probiotics) treatment will be recommended in insectaries considering both the beneficial effects on mass reared insects and its general safety for insectary workers and for environment.


Asunto(s)
Ceratitis capitata/inmunología , Ceratitis capitata/efectos de la radiación , Dieta , Inmunidad/efectos de los fármacos , Infertilidad Masculina/inmunología , Control Biológico de Vectores , Probióticos/farmacología , Estrés Fisiológico/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Ceratitis capitata/genética , Radioisótopos de Cobalto , Femenino , Vuelo Animal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Inmunidad/genética , Inmunidad/efectos de la radiación , Infertilidad Masculina/genética , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Masculino , Pupa/efectos de los fármacos , Estadística como Asunto , Estrés Fisiológico/genética
10.
Int J Mol Sci ; 22(14)2021 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-34298925

RESUMEN

The Linear-No-Threshold (LNT) model predicts a dose-dependent linear increase in cancer risk. This has been supported by biological and epidemiological studies at high-dose exposures. However, at low-doses (LDR ≤ 0.1 Gy), the effects are more elusive and demonstrate a deviation from linearity. In this study, the effects of LDR on the development and progression of mammary cancer in FVB/N-Tg(MMTVneu)202Mul/J mice were investigated. Animals were chronically exposed to total doses of 10, 100, and 2000 mGy via tritiated drinking water, and were assessed at 3.5, 6, and 8 months of age. Results indicated an increased proportion of NK cells in various organs of LDR exposed mice. LDR significantly influenced NK and T cell function and activation, despite diminishing cell proliferation. Notably, the expression of NKG2D receptor on NK cells was dramatically reduced at 3.5 months but was upregulated at later time-points, while the expression of NKG2D ligand followed the opposite trend, with an increase at 3.5 months and a decrease thereafter. No noticeable impact was observed on mammary cancer development, as measured by tumor load. Our results demonstrated that LDR significantly influenced the proportion, proliferation, activation, and function of immune cells. Importantly, to the best of our knowledge, this is the first report demonstrating that LDR modulates the cross-talk between the NKG2D receptor and its ligands.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/radioterapia , Inmunidad/efectos de la radiación , Animales , Neoplasias de la Mama/metabolismo , Proliferación Celular/efectos de la radiación , Femenino , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/efectos de la radiación , Ligandos , Ratones , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Dosis de Radiación , Linfocitos T/metabolismo , Linfocitos T/efectos de la radiación , Regulación hacia Arriba/efectos de la radiación
11.
Front Immunol ; 12: 680503, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34079557

RESUMEN

In the past decade, radiation therapy (RT) entered the era of personalized medicine, following the striking improvements in radiation delivery and treatment planning optimization, and in the understanding of the cancer response, including the immunological response. The next challenge is to identify the optimal radiation regimen(s) to induce a clinically relevant anti-tumor immunity response. Organs at risks and the tumor microenvironment (e.g. endothelial cells, macrophages and fibroblasts) often limit the radiation regimen effects due to adverse toxicities. Here, we reviewed how RT can modulate the immune response involved in the tumor control and side effects associated with inflammatory processes. Moreover, we discussed the versatile roles of tumor microenvironment components during RT, how the innate immune sensing of RT-induced genotoxicity, through the cGAS-STING pathway, might link the anti-tumor immune response, radiation-induced necrosis and radiation-induced fibrosis, and how a better understanding of the switch between favorable and deleterious events might help to define innovative approaches to increase RT benefits in patients with cancer.


Asunto(s)
Inmunidad/efectos de la radiación , Radioterapia/efectos adversos , Animales , Efecto Espectador/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Humanos , Proteínas de la Membrana/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/radioterapia , Nucleotidiltransferasas/metabolismo , Especificidad de Órganos/inmunología , Especificidad de Órganos/efectos de la radiación , Radiación Ionizante , Radioterapia/métodos , Transducción de Señal/efectos de la radiación , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación
12.
J Nanobiotechnology ; 19(1): 142, 2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34001148

RESUMEN

BACKGROUND: Therapeutic tumor vaccine (TTV) that induces tumor-specific immunity has enormous potentials in tumor treatment, but high heterogeneity and poor immunogenicity of tumor seriously impair its clinical efficacy. Herein, a novel NIR responsive tumor vaccine in situ (HA-PDA@IQ/DOX HG) was prepared by integrating hyaluronic acid functionalized polydopamine nanoparticles (HA-PDA NPs) with immune adjuvants (Imiquimod, IQ) and doxorubicin (DOX) into thermal-sensitive hydrogel. RESULTS: HA-PDA@IQ NPs with high photothermal conversion efficiency (41.2%) and T1-relaxation efficiency were using HA as stabilizer by the one-pot oxidative polymerization. Then, HA-PDA@IQ loaded DOX via π-π stacking and mixed with thermal-sensitive hydrogel to form the HA-PDA@IQ/DOX HG. The hydrogel-confined delivery mode endowed HA-PDA@IQ/DOX NPs with multiple photothermal ablation performance once injection upon NIR irradiation due to the prolonged retention in tumor site. More importantly, this mode enabled HA-PDA@IQ/DOX NPs to promote the DC maturation, memory T cells in lymphatic node as well as cytotoxic T lymphocytes in spleen. CONCLUSION: Taken together, the HA-PDA@IQ/DOX HG could be served as a theranostic tumor vaccine for complete photothermal ablation to trigger robust antitumor immune responses.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Quimioterapia/métodos , Inmunidad/efectos de los fármacos , Inmunidad/efectos de la radiación , Terapia Fototérmica/métodos , Doxorrubicina/administración & dosificación , Ácido Hialurónico , Hidrogeles , Indoles/química , Nanopartículas/administración & dosificación , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Polímeros/administración & dosificación , Polímeros/química
13.
J Exp Clin Cancer Res ; 40(1): 161, 2021 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-33964942

RESUMEN

BACKGROUND: Improvement of radiotherapy efficacy requires better insight in the dynamic responses that occur during irradiation. Here, we aimed to identify the molecular responses that are triggered during clinically applied fractionated irradiation. METHODS: Gene expression analysis was performed by RNAseq or microarray analysis of cancer cells or xenograft tumors, respectively, subjected to 3-5 weeks of 5 × 2 Gy/week. Validation of altered gene expression was performed by qPCR and/or ELISA in multiple cancer cell lines as well as in pre- and on-treatment biopsies from esophageal cancer patients ( NCT02072720 ). Targeted protein inhibition and CRISPR/Cas-induced gene knockout was used to analyze the role of type I interferons and cGAS/STING signaling pathway in the molecular and cellular response to fractionated irradiation. RESULTS: Gene expression analysis identified type I interferon signaling as the most significantly enriched biological process induced during fractionated irradiation. The commonality of this response was confirmed in all irradiated cell lines, the xenograft tumors and in biopsies from esophageal cancer patients. Time-course analyses demonstrated a peak in interferon-stimulated gene (ISG) expression within 2-3 weeks of treatment. The response was accompanied by a variable induction of predominantly interferon-beta and/or -lambda, but blocking these interferons did not affect ISG expression induction. The same was true for targeted inhibition of the upstream regulatory STING protein while knockout of STING expression only delayed the ISG expression induction. CONCLUSIONS: Collectively, the presented data show that clinically applied fractionated low-dose irradiation can induce a delayed type I interferon response that occurs independently of interferon expression or STING signaling. These findings have implications for current efforts that aim to target the type I interferon response for cancer treatment.


Asunto(s)
Neoplasias Esofágicas/radioterapia , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Interferón Tipo I/genética , Proteínas de la Membrana/genética , Animales , Astrocitoma/genética , Astrocitoma/inmunología , Astrocitoma/metabolismo , Astrocitoma/radioterapia , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/radioterapia , Fraccionamiento de la Dosis de Radiación , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/inmunología , Neoplasias Esofágicas/metabolismo , Femenino , Células HT29 , Humanos , Inmunidad/efectos de la radiación , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Front Immunol ; 12: 646384, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054811

RESUMEN

Inadequate sustained immune activation and tumor recurrence are major limitations of radiotherapy (RT), sustained and targeted activation of the tumor microenvironment can overcome this obstacle. Here, by two models of a primary rat breast cancer and cell co-culture, we demonstrated that valproic acid (VPA) and its derivative (HPTA) are effective immune activators for RT to inhibit tumor growth by inducing myeloid-derived macrophages and polarizing them toward the M1 phenotype, thus elevate the expression of cytokines such as IL-12, IL-6, IFN-γ and TNF-α during the early stage of the combination treatment. Meanwhile, activated CD8+ T cells increased, angiogenesis of tumors is inhibited, and the vasculature becomes sparse. Furthermore, it was suggested that VPA/HPTA can enhance the effects of RT via macrophage-mediated and macrophage-CD8+ T cell-mediated anti-tumor immunity. The combination of VPA/HPTA and RT treatment slowed the growth of tumors and prolong the anti-tumor effect by continuously maintaining the activated immune response. These are promising findings for the development of new effective, low-cost concurrent cancer therapy.


Asunto(s)
Neoplasias de la Mama/radioterapia , Linfocitos T CD8-positivos/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Tolerancia a Radiación/efectos de los fármacos , Ácido Valproico/farmacología , Animales , Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Humanos , Inmunidad/efectos de los fármacos , Inmunidad/inmunología , Inmunidad/efectos de la radiación , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Tolerancia a Radiación/inmunología , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Transducción de Señal/efectos de la radiación , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación , Ácido Valproico/química
15.
Radiat Res ; 195(6): 522-540, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33826741

RESUMEN

Brain metastases develop in over 60% of advanced melanoma patients and negatively impact quality of life and prognosis. In a murine melanoma model, we previously showed that an in situ vaccination (ISV) regimen, combining radiation treatment and intratumoral (IT) injection of immunocytokine (IC: anti-GD2 antibody fused to IL2), along with the immune checkpoint inhibitor anti-CTLA-4, robustly eliminates peripheral flank tumors but only has modest effects on co-occurring intracranial tumors. In this study, we investigated the ability of low-dose radiation to the brain to potentiate anti-tumor immunity against a brain tumor when combined with ISV + anti-CTLA-4. B78 (GD2+, immunologically "cold") melanoma tumor cells were implanted into the flank and the right striatum of the brain in C57BL/6 mice. Flank tumors (50-150 mm3) were treated following a previously optimized ISV regimen [radiation (12 Gy × 1, treatment day 1), IT-IC (50 µg daily, treatment days 6-10), and anti-CTLA-4 (100 µg, treatment days 3, 6, 9)]. Mice that additionally received whole-brain radiation treatment (WBRT, 4 Gy × 1) on day 15 demonstrated significantly increased survival compared to animals that received ISV + anti-CTLA-4 alone, WBRT alone or no treatment (control) (P < 0.001, log-rank test). Timing of WBRT was critical, as WBRT administration on day 1 did not significantly enhance survival compared to ISV + anti-CTLA-4, suggesting that the effect of WBRT on survival might be mediated through immune modulation and not just direct tumor cell cytotoxicity. Modest increases in T cells (CD8+ and CD4+) and monocytes/macrophages (F4/80+) but no changes in FOXP3+ regulatory T cells (Tregs), were observed in brain melanoma tumors with addition of WBRT (on day 15) to ISV + anti-CTLA-4. Cytokine multiplex immunoassay revealed distinct changes in both intracranial melanoma and contralateral normal brain with addition of WBRT (day 15) to ISV + anti-CTLA-4, with notable significant changes in pro-inflammatory (e.g., IFNγ, TNFα and LIX/CXCL5) and suppressive (e.g., IL10, IL13) cytokines as well as chemokines (e.g., IP-10/CXCL10 and MIG/CXCL9). We tested the ability of the alkylphosphocholine analog, NM600, to deliver immunomodulatory radiation to melanoma brain tumors as a targeted radionuclide therapy (TRT). Yttrium-86 (86Y) chelated to NM600 was delivered intravenously by tail vein to mice harboring flank and brain melanoma tumors, and PET imaging demonstrated specific accumulation up to 72 h at each tumor site (∼12:1 brain tumor/brain and ∼8:1 flank tumor/muscle). When NM600 was chelated to therapeutic ß-particle-emitting 90Y and administered on treatment day 13, T-cell infiltration and cytokine profiles were altered in melanoma brain tumor, like that observed for WBRT. Overall, our results demonstrate that addition of low-dose radiation, timed appropriately with ISV administration to tumors outside the brain, significantly increases survival in animals co-harboring melanoma brain tumors. This observation has potentially important translational implications as a treatment strategy for increasing the response of tumors in the brain to systemically administered immunotherapies.


Asunto(s)
Neoplasias Encefálicas/inmunología , Inmunidad/efectos de la radiación , Melanoma Experimental/inmunología , Vacunación , Animales , Neoplasias Encefálicas/prevención & control , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunidad/efectos de los fármacos , Melanoma Experimental/prevención & control , Ratones , Ratones Endogámicos C57BL , Proteína Tumoral Controlada Traslacionalmente 1
16.
Hum Immunol ; 82(6): 446-454, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33812705

RESUMEN

Ionizing radiation, including X and gamma rays, are used for various purposes such as; medicine, nuclear power, research, manufacturing, food preservation and construction. Furthermore, people are also exposed to ionizing radiation from their workplace or the environment. Apart from DNA fragmentation resulting in apoptosis, several additional mechanisms have been proposed to describe how radiation can alter human cell functions. Ionizing radiation may alter immune responses, which are the main cause of human disorders. Toll like receptors (TLRs) are important human innate immunity receptors which participate in several immune and non-immune cell functions including, induction of appropriate immune responses and immune related disorders. Based on the role played by ionizing radiation on human cell systems, it has been hypothesized that radiation may affect immune responses. Therefore, the main aim of this review article is to discuss recent information regarding the effects of ionizing radiation on TLRs and their related disorders.


Asunto(s)
Enfermedades del Sistema Inmune/inmunología , Inmunidad/efectos de la radiación , Receptores Toll-Like/metabolismo , Animales , Humanos , Inmunidad Innata , Inmunomodulación , Radiación Ionizante
17.
Cell Cycle ; 19(24): 3595-3607, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33283623

RESUMEN

Microwave ablation (MWA) has been widely used in the treatment of solid tumors. Studies have been less conducted on the efficacy of MWA used with cell immunotherapy in treating hepatocellular carcinoma (HCC). The current study aimed at exploring the efficacy of MWA in combination with cell immunotherapy in treating HCC. Hepa1-6 HCC mice were treated by MWA, blockade, or the combined therapy (MWA used with blockade), or left untreated. Survival rates of the mice were plotted by Kaplan-Meier Curve, followed by log-rank test. 25 days after the operation, surviving mice were monitored for tumor recurrence, and tumor volumes were calculated every 5 days. Immunohistochemistry and flow cytometry were performed to detect the numbers of CD4+ and CD8+ cells in the tumors and spleens of mice. The expressions of related cytokines were detected and measured by ELISPOT and ELISA. The results showed that MWA combined with anti-PD-1/anti-CTLA-4 not only increased the survival time, protected the mice against tumor recurrence, but also enhanced the intratumoral infiltration of cytotoxic T lymphocyte and systemic T-cell immune responses induced by MWA through activation of synergistically specific antitumor immunity. In addition, the combined therapy increased T-helper 1 cell (Th1-type) cytokines, but reduced Th2-type cytokines, resulting in the polarization of Th1 cells. T-cell immune responses of HCC cells were activated by MWA. In addition, the combined therapy of MWA and anti-PD-1/anti-CTLA-4 induced Th1-type immune response, and showed specific antitumor immunity.


Asunto(s)
Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/terapia , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunidad/efectos de la radiación , Inmunoterapia/métodos , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/terapia , Microondas/uso terapéutico , Terapia por Radiofrecuencia/métodos , Animales , Antígeno CTLA-4/antagonistas & inhibidores , Terapia Combinada/métodos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Tasa de Supervivencia , Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología , Resultado del Tratamiento , Carga Tumoral/efectos de la radiación
18.
Int J Mol Sci ; 21(22)2020 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-33238631

RESUMEN

Radiotherapy is a major modality used to combat a wide range of cancers. Classical radiobiology principles categorize ionizing radiation (IR) as a direct cytocidal therapeutic agent against cancer; however, there is an emerging appreciation for additional antitumor immune responses generated by this modality. A more nuanced understanding of the immunological pathways induced by radiation could inform optimal therapeutic combinations to harness radiation-induced antitumor immunity and improve treatment outcomes of cancers refractory to current radiotherapy regimens. Here, we summarize how radiation-induced DNA damage leads to the activation of a cytosolic DNA sensing pathway mediated by cyclic GMP-AMP (cGAMP) synthase (cGAS) and stimulator of interferon genes (STING). The activation of cGAS-STING initiates innate immune signaling that facilitates adaptive immune responses to destroy cancer. In this way, cGAS-STING signaling bridges the DNA damaging capacity of IR with the activation of CD8+ cytotoxic T cell-mediated destruction of cancer-highlighting a molecular pathway radiotherapy can exploit to induce antitumor immune responses. In the context of radiotherapy, we further report on factors that enhance or inhibit cGAS-STING signaling, deleterious effects associated with cGAS-STING activation, and promising therapeutic candidates being investigated in combination with IR to bolster immune activation through engaging STING-signaling. A clearer understanding of how IR activates cGAS-STING signaling will inform immune-based treatment strategies to maximize the antitumor efficacy of radiotherapy, improving therapeutic outcomes.


Asunto(s)
Anomalías Inducidas por Radiación/genética , Inmunidad/genética , Proteínas de la Membrana/genética , Neoplasias/genética , Nucleotidiltransferasas/genética , Anomalías Inducidas por Radiación/inmunología , Anomalías Inducidas por Radiación/patología , Daño del ADN/inmunología , Daño del ADN/efectos de la radiación , Humanos , Inmunidad/inmunología , Inmunidad/efectos de la radiación , Inmunidad Innata/efectos de la radiación , Proteínas de la Membrana/inmunología , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/radioterapia , Nucleotidiltransferasas/inmunología , Transducción de Señal/efectos de la radiación
19.
PLoS One ; 15(10): e0241375, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33119684

RESUMEN

Humanized mouse models are used extensively in research involving human pathogens and diseases. However, most of these models require preconditioning. Radio-active sources have been used routinely for this purpose but safety issues have motivated researchers to transition to chemical or X-ray based preconditioning. In this study, we directly compare 350 kV X-ray and Cs-137 low-dose precondition of NOG mice before human stem cell transplantation. Based on flow cytometry data, we found that engraftment of human cells into the mouse bone marrow was similar between radiation sources. Likewise, human engraftment in the peripheral blood was comparable between Cs-137 and three different X-ray doses with equal chimerization kinetics. In primary lymphoid organs such as the thymus and lymph nodes, and spleen, liver and lung, human-to-mouse chimerization was also comparable between irradiation sources. Development of different CD4 and CD8 T cells as well as these cells' maturation stages, i.e. from naïve to effector and memory subsets were generally analogous. Based on our results, we conclude that there are no discernable differences between the two sources in the low-dose spectrum investigated. However, while we encourage the transition to X-ray-based sources, we recommend all research groups to consider technical specifications and dose-finding studies.


Asunto(s)
Radioisótopos de Cesio , Inmunidad/efectos de la radiación , Rayos X , Animales , Trasplante de Médula Ósea , Femenino , Humanos , Cinética , Ganglios Linfáticos/inmunología , Ratones , Fenotipo , Timo/inmunología
20.
Int J Radiat Biol ; 96(11): 1466-1472, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32910717

RESUMEN

PURPOSE: Long-term exposure to low dose radiation may trigger immune response and stimulate hormesis. Interleukin-2 (IL-2) and interleukin-2 receptor (IL-2R) play a crucial role in immune function. We aimed to explore the possible association of IL-2 and IL-2R gene polymorphisms with low dose radiation exposure, as well as the relationship with IL-2 gene expression in people residing in areas with a high background radiation in Yangjiang, China. MATERIALS AND METHODS: We recruited and assigned 54 native men residing in Yangxi County, Yangjiang city to the high natural background radiation (HNBR) group, and 53 native men residing in Hengpi County, Enping city to the control area (CA) group. All the participants wore a thermoluminescent dosimeter (TLD) for 90 days, and answered questionnaires. The serum levels of IL2, IL4, IL5, sIL2R, and tumor growth factor (TGF), and expression levels of IL2RA, IL2RB, IL2RG, and IL2 were also analyzed. Additionally, we tested 10 polymorphic loci associated with the IL-2 gene. RESULTS: The annual effective radiation doses in the HNBR and CA groups were 6.24 mSv y-1 and 1.95 mSv y-1, respectively. After adjusting for potential confounding factors, the serum levels of IL-2 and IL-5 were higher in the HNBR group than the CA group (p < .05), while the serum level of TGFß was lower in the HNBR group (p < .05). The IL-2 gene mRNA expression level was higher in the HNBR group than the CA group (p < .05). The IL-2RB rs76206423 AA allele showed significant variations in the HNBR group (p = .0381). CONCLUSIONS: Long-term exposure to low dose radiation may enhance immune function, and IL-2RB rs76206423 may be related to the expression of IL-2 by other coding variants. Moreover, our data provide a better understanding of the molecular mechanism of the immune response to low dose radiation.


Asunto(s)
Radiación de Fondo/efectos adversos , Inmunidad/efectos de la radiación , Interleucina-2/genética , Polimorfismo de Nucleótido Simple/efectos de la radiación , Receptores de Interleucina-2/genética , China , Femenino , Regulación de la Expresión Génica/efectos de la radiación , Genotipo , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA