Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 693
Filtrar
1.
Cancer Med ; 10(21): 7475-7491, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34626092

RESUMEN

BACKGROUND: We discovered that pure positive electrostatic charges (PECs) have an intrinsic suppressive effect on the proliferation and metabolism of invasive cancer cells (cell lines and animal models) without affecting normal tissues. METHODS: We interacted normal and cancer cell lines and animal tumors with PECs by connecting a charged patch to cancer cells and animal tumors. many biochemical, molecular and radiological assays were carried out on PEC treated and control samples. RESULTS: Correlative interactions between electrostatic charges and cancer cells contain critical unknown factors that influence cancer diagnosis and treatment. Different types of cell analyses prove PEC-based apoptosis induction in malignant cell lines. Flowcytometry and viability assay depict selective destructive effects of PEC on malignant breast cancer cells. Additionally, strong patterns of pyknotic apoptosis, as well as downregulation of proliferative-associated proteins (Ki67, CD31, and HIF-1α), were observed in histopathological and immunohistochemical patterns of treated mouse malignant tumors, respectively. Quantitative real-time polymerase chain reaction results demonstrate up/down-regulated apoptotic/proliferative transcriptomes (P21, P27, P53/CD34, integrin α5, vascular endothelial growth factor, and vascular endothelial growth factor receptor) in treated animal tumors. Expression of propidium iodide in confocal microscopy images of treated malignant tissues was another indication of the destructive effects of PECs on such cells. Significant tumor size reduction and prognosis improvement were seen in over 95% of treated mouse models with no adverse effects on normal tissues. CONCLUSION: We discovered that pure positive electrostatic charges (PECs) have an intrinsic suppressive effect on the proliferation and metabolism of invasive cancer cells (cell lines and animal models) without affecting normal tissues. The findings were statistically and observationally significant when compared to radio/chemotherapy-treated mouse models. As a result, this nonionizing radiation may be used as a practical complementary approach with no discernible side effects after passing future human model studies.


Asunto(s)
Proliferación Celular , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/terapia , Electricidad Estática , Animales , Apoptosis , Línea Celular Tumoral , Modelos Animales de Enfermedad , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/radioterapia , Necrosis , Clasificación del Tumor
2.
Biomed Environ Sci ; 34(9): 693-704, 2021 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-34530959

RESUMEN

OBJECTIVE: The aim of this study was to explore the effects of 2-hexyl-4-pentylenic acid (HPTA) in combination with radiotherapy (RT) on distant unirradiated breast tumors. METHODS: Using a rat model of chemical carcinogen (7,12-dimethylbenz[a]anthracene,DMBA)-induced breast cancer, tumor volume was monitored and treatment response was evaluated by performing HE staining, immunohistochemistry, immunofluorescence, qRT-PCR, and western blot analyses. RESULTS: The results demonstrated that HPTA in combination with RT significantly delayed the growth of distant, unirradiated breast tumors. The mechanism of action included tumor-associated macrophage (TAM) infiltration into distant tumor tissues, M1 polarization, and inhibition of tumor angiogenesis by IFN-γ. CONCLUSION: The results suggest that the combination of HPTA with RT has an abscopal effect on distant tumors via M1-polarized TAMs, and HPTA may be considered as a new therapeutic for amplifying the efficacy of local RT for non-targeted breast tumors.The graphical abstract was available in the web of www.besjournal.com.


Asunto(s)
Antineoplásicos/uso terapéutico , Ácidos Grasos Insaturados/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/radioterapia , Animales , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Terapia Combinada , Citocinas/inmunología , Ácidos Grasos Insaturados/farmacología , Femenino , Neoplasias Mamarias Experimentales/inmunología , Ratas , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/efectos de la radiación
3.
Dokl Biochem Biophys ; 498(1): 159-164, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34189642

RESUMEN

The combined effect of the irradiation with a proton pencil scanning beam (PBS) at a total dose of 80 Gy and neutron radiation at a dose of 5 Gy on the growth of solid Ehrlich ascites carcinoma (EAC) and the remote effects in tumor-bearing mice was studied. Combined irradiation of mice with neutrons before and after irradiation with PBS, as well as irradiation only with PBS, effectively suppressed the growth of solid EAC within 1 month. In terms of the frequency and severity of radiation-induced skin reactions of mice observed 15-40 days after therapy, neutron irradiation after the irradiation with PBS showed better values of these parameters as compared to only PBS; however, exposure to neutrons before PBS was more damaging as compared to the other two options. It was also shown that the tumor relapse rate in the groups of animals with combined irradiation was higher, and the total lifespan was lower than the group of mice irradiated with PBS alone.


Asunto(s)
Carcinoma de Ehrlich/radioterapia , Neoplasias Mamarias Experimentales/radioterapia , Neutrones/uso terapéutico , Terapia de Protones , Animales , Carcinoma de Ehrlich/metabolismo , Carcinoma de Ehrlich/patología , Femenino , Masculino , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Tasa de Supervivencia , Resultado del Tratamiento
4.
Int J Biol Sci ; 17(3): 689-701, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33767581

RESUMEN

Although germline mutations in BRCA1 highly predispose women towards breast and ovarian cancer, few substantial improvements in preventing or treating such cancers have been made. Importantly, BRCA1 function is closely associated with DNA damage repair, which is required for genetic stability. Here, we examined the efficacy of radiotherapy, assessing the accumulation of genetic instabilities, in the treatment of BRCA1-associated breast cancer using a Brca1-mutant mouse model. Treatment of Brca1-mutant tumor-engrafted mice with X-rays reduced tumor progression by 27.9% compared with untreated controls. A correlation analysis of irradiation responses and biomarker profiles in tumors at baseline identified differences between responders and non-responders at the protein level (pERα, pCHK2, p53, and EpCAM) and at the SOX2 target expression level. We further demonstrated that combined treatment of Brca1-mutant mammary tumors with irradiation and AZD2281, which inhibits PARP, significantly reduced tumor progression and extended survival. Our findings enhance the understanding of DNA damage and biomarker responses in BRCA1-associated mammary tumors and provide preclinical evidence that radiotherapy with synthetic DNA damage is a potential strategy for the therapeutic management of BRCA1-associated breast cancer.


Asunto(s)
Genes BRCA1 , Neoplasias Mamarias Experimentales/radioterapia , Ftalazinas/uso terapéutico , Piperazinas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Animales , Biomarcadores de Tumor/metabolismo , Quimioradioterapia/métodos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Células MCF-7 , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Noqueados
5.
Anticancer Res ; 41(1): 55-70, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33419799

RESUMEN

BACKGROUND/AIM: Our understanding of cancer risk from neutron exposure is limited. We aimed to reveal the characteristics of mammary carcinomas induced by neutrons. MATERIALS AND METHODS: Mammary carcinomas obtained from female Sprague-Dawley rats irradiated at 7 weeks of age with 0.97 Gy neutrons or 4 Gy γ-rays and from non-irradiated rats were classified into luminal and non-luminal subtypes by immunohistochemistry. Their mutational landscapes were determined by whole-exome sequencing. RESULTS: Neutrons significantly raised the incidence of luminal mammary carcinomas over the non-luminal subtype. Somatic mutations were identified in cancer genes involved in several signalling pathways, including Keap1/Nrf2, Pi3k/Akt and Wnt/ß-catenin. Focal copy-number losses involving cancer genes were observed mainly in carcinomas from the irradiated rats. CONCLUSION: Neutrons increase the incidence of luminal mammary carcinomas, probably through gene mutations similar to those found in human breast cancers, and focal copy-number losses including cancer genes that are characteristics of radiation-induced mammary carcinomas.


Asunto(s)
Variaciones en el Número de Copia de ADN/efectos de la radiación , Exoma , Neoplasias Mamarias Experimentales/genética , Mutación/efectos de la radiación , Radiación Ionizante , Animales , Biopsia , Biología Computacional/métodos , Metilación de ADN , Análisis Mutacional de ADN , Femenino , Humanos , Mutación INDEL , Inmunohistoquímica , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/radioterapia , Neoplasias Inducidas por Radiación/genética , Neoplasias Inducidas por Radiación/patología , Ratas , Secuenciación del Exoma
6.
J Mater Chem B ; 9(6): 1625-1637, 2021 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-33475658

RESUMEN

Radiotherapy occupies an essential position in curing and palliating a wide range of solid tumors based on DNA damage responses to eradicate cancer cells. However, the tumor microenvironment generally exhibits the characteristics of hypoxia and glutathione overexpression, which play a critical role in radioresistance, to prevent irreparable breaks to DNA and necrocytosis of cancer cells. Herein, polyethylene glycol (PEG) functionalized manganese ferrite nanoparticles (MnFe2O4-PEG) are designed to enable self-sufficiency of oxygen by continuously catalyzing the decomposition of endogenous hydrogen peroxide. Simultaneously, the nano-platform can consume GSH to reduce the loss of reactive oxygen species in radiotherapy and achieve better therapeutic effects at the cellular and animal levels. In addition, the MnFe2O4-PEG could act as an optimal T1- and T2-weighted contrast medium for tumor-specific magnetic resonance imaging. This work proposes a systematically administered radiosensitizer that can selectively reside in tumor sites via the enhanced permeability and retention effect to relieve hypoxia and reduce GSH concentration, combined with dual-mode magnetic resonance imaging, achieving precise and effective image-guided tumor therapy.


Asunto(s)
Neoplasias de la Mama/radioterapia , Compuestos Férricos/farmacología , Imagen por Resonancia Magnética , Compuestos de Manganeso/farmacología , Polietilenglicoles/farmacología , Hipoxia Tumoral/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Femenino , Compuestos Férricos/química , Humanos , Neoplasias Mamarias Experimentales/radioterapia , Compuestos de Manganeso/química , Ratones , Ratones Endogámicos BALB C , Tamaño de la Partícula , Técnicas Fotoacústicas , Polietilenglicoles/química , Propiedades de Superficie , Microambiente Tumoral/efectos de los fármacos
7.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-921320

RESUMEN

Objective@#The aim of this study was to explore the effects of 2-hexyl-4-pentylenic acid (HPTA) in combination with radiotherapy (RT) on distant unirradiated breast tumors.@*Methods@#Using a rat model of chemical carcinogen (7,12-dimethylbenz[a]anthracene,DMBA)-induced breast cancer, tumor volume was monitored and treatment response was evaluated by performing HE staining, immunohistochemistry, immunofluorescence, qRT-PCR, and western blot analyses.@*Results@#The results demonstrated that HPTA in combination with RT significantly delayed the growth of distant, unirradiated breast tumors. The mechanism of action included tumor-associated macrophage (TAM) infiltration into distant tumor tissues, M1 polarization, and inhibition of tumor angiogenesis by IFN-γ.@*Conclusion@#The results suggest that the combination of HPTA with RT has an abscopal effect on distant tumors


Asunto(s)
Animales , Femenino , Ratas , Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de la radiación , Terapia Combinada , Citocinas/inmunología , Ácidos Grasos Insaturados/uso terapéutico , Neoplasias Mamarias Experimentales/radioterapia , Macrófagos Asociados a Tumores/efectos de la radiación
8.
Naunyn Schmiedebergs Arch Pharmacol ; 393(2): 253-262, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31522240

RESUMEN

Heat shock protein 90 (HSP90) inhibitors are considered as new radiosensitizing agents. PU-H71, a novel HSP90 inhibitor, is under evaluation for the treatment of advanced cancer. It is however not known whether PU-H71 alters radiosensitivity of metastatic breast cancer. Hence, we here evaluated mechanisms of possible anti-tumoral and radiosensitizing effects of PU-H71 on breast carcinoma cells metastasized to vital organs such as the liver and brain. The effect of PU-H71 on proliferation of breast carcinoma cells was determined using 4T1 cells and its brain (4TBM), liver (4TLM), and heart (4THM) metastatic subsets as well as non-metastatic 67NR cells. Changes in radiation sensitivity were determined by clonogenic assays. Changes in client proteins and levels of angiogenic and inflammatory mediators from these cancer cell cultures and ex vivo cultures were detected. PU-H71 alone inhibited ERK1/2, p38, and Akt activation and reduced N-cadherin and HER2 which further documented the anti-tumoral effects of PU-H71. The combination of PU-H71 and radiotherapy induced cytotoxic effect than PU-H71 alone, and PU-H71 showed a radiosensitizing effect in vitro. On the other hand, PU-H71 and radiation co-treatment increased p38 phosphorylation which is one of the hallmarks of inflammatory response. Accordingly, IL-6 secretion was increased following PU-H71 and radiotherapy co-treatment ex vivo. Levels of angiogenic and inflammatory factors such as MIP-2, SDF-1, and VEGF were increased under in vitro conditions but not under ex vivo conditions. These results demonstrated for the first time that PU-H71 enhances therapeutic effects of radiotherapy especially in highly metastatic breast carcinoma but a possible increase in inflammatory response should also be considered.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzodioxoles/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/radioterapia , Purinas/uso terapéutico , Tolerancia a Radiación/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Benzodioxoles/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Quimiocina CXCL2/metabolismo , Femenino , Mediadores de Inflamación , Interleucina-6/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Purinas/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Clin Cancer Res ; 26(4): 910-921, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31757878

RESUMEN

PURPOSE: Radiotherapy (RT) is frequently used for local control of solid tumors using equal dose per fraction. Recently, single high-dose radiation has been used for ablation of solid tumors. In this report, we provide a novel immunological basis for radiation dose fractionation consisting of a single high-dose radiotherapy, followed by postablation modulation (PAM) with four daily low-dose fractions (22 Gy + 0.5 Gy × 4) to reprogram the tumor microenvironment by diminishing immune suppression, enabling infiltration of effector cells and increasing efficacy of tumor control. EXPERIMENTAL DESIGN: Palpable 3LL and 4T1 tumors in C57Bl/6 and Balb/c mice were irradiated with the Small-Animal Radiation Research Platform irradiator, and tumor growth and survival were monitored. Immunomodulation of tumor and immune cells in vitro and in vivo characterization of tumor-infiltrating immune effector cells were performed by FACS. For systemic application of PAM-RT, whole-lung irradiation was administered in 4T1-bearing Balb/c mice. RESULTS: We report significant tumor growth delays and increased survival in 3LL tumor-bearing mice with PAM. Primary tumor PAM-RT increased infiltration of immune effector cells and decreased Treg in irradiated tumors and secondary lymphoid organs. In a model of murine metastatic breast cancer (4T1), we demonstrated that systemic PAM-RT to the whole lung, 12 days after primary tumor ablative radiotherapy, increased survival with suppression of pulmonary metastases. CONCLUSIONS: We provide a novel immunologic basis for radiation dose fractionation consisting of a single high dose of radiotherapy followed by daily low-dose PAM-RT fractionation to improve the immunogenic potential of ablative radiotherapy.


Asunto(s)
Carcinoma Pulmonar de Lewis/radioterapia , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Experimentales/radioterapia , Microambiente Tumoral/inmunología , Técnicas de Ablación/métodos , Animales , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/cirugía , Línea Celular Tumoral , Fraccionamiento de la Dosis de Radiación , Femenino , Inmunomodulación/efectos de la radiación , Linfocitos Infiltrantes de Tumor/efectos de la radiación , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/cirugía , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Tasa de Supervivencia , Microambiente Tumoral/efectos de la radiación
10.
Clin Cancer Res ; 26(4): 945-956, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31694834

RESUMEN

PURPOSE: There is growing interest in combinations of immunogenic radiotherapy (RT) and immune checkpoint blockade, but clinical responses are still limited. Therefore, we tested the triple therapy with an inhibitor of the indoleamine 2,3-dioxygenase pathway, which like immune checkpoints, downregulates the antitumor immune response. EXPERIMENTAL DESIGN: Triple treatment with hypofractionated RT (hRT) + anti-PD-1 antibody (αPD1) + indoximod was compared with the respective mono- and dual therapies in two syngeneic mouse models. RESULTS: The tumors did not regress following treatment with hRT + αPD1. The αPD1/indoximod combination was not effective at all. In contrast, triple treatment induced rapid, marked tumor regression, even in mice with a large tumor. The effects strongly depended on CD8+ T cells and partly on natural killer (NK) cells. Numbers and functionality of tumor-specific CD8+ T cells and NK cells were increased, particularly early during treatment. However, after 2.5-3 weeks, all large tumors relapsed, which was accompanied by increased apoptosis of tumor-infiltrating lymphocytes associated with a non-reprogrammable state of exhaustion, terminal differentiation, and increased activation-induced cell death, which could not be prevented by indoximod in these aggressive tumor models. Some mice with a smaller tumor were cured. Reirradiation during late regression (day 12), but not after relapse, cured almost all mice with a large B16-CD133 tumor, and strongly delayed relapse in the less immunogenic 4T1 model, depending on CD8+ T cells. CONCLUSIONS: Our findings may serve as a rationale for the clinical evaluation of this triple-combination therapy in patients with solitary or oligometastatic tumors in the neoadjuvant or the definitive setting.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Asesinas Naturales/inmunología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/radioterapia , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/radioterapia , Triptófano/análogos & derivados , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de la radiación , Línea Celular Tumoral , Quimioradioterapia , Femenino , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/efectos de la radiación , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Hipofraccionamiento de la Dosis de Radiación , Tasa de Supervivencia , Triptófano/farmacología
11.
J Control Release ; 314: 1-11, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31626860

RESUMEN

Radioimmunotherapy (RIT) aims to deliver a high radiation dose to cancer cells, while minimizing the exposure of normal cells. Typically, monoclonal antibodies are used to target the radionuclides to cancer cell surface antigens. However, antibodies face limitations due to their poor tumor penetration and suboptimal pharmacokinetics, while the expression of their target on the cancer cell surface may be gradually lost. In addition, most antigens are expressed in a limited number of tumor types. To circumvent these problems, we developed a Nanobody (Nb)-based RIT against a prominent stromal cell (stromal-targeting radioimmunotherapy or STRIT) present in nearly all tumors, the tumor-associated macrophage (TAM). Macrophage Mannose Receptor (MMR) functions as a stable molecular target on TAM residing in hypoxic areas, further allowing the delivery of a high radiation dose to the more radioresistant hypoxic tumor regions. Since MMR expression is not restricted to TAM, we first optimized a strategy to block extra-tumoral MMR to prevent therapy-induced toxicity. A 100-fold molar excess of unlabeled bivalent Nb largely blocks extra-tumoral binding of 177Lu-labeled anti-MMR Nb and prevents toxicity, while still allowing the intra-tumoral binding of the monovalent Nb. Interestingly, three doses of 177Lu-labeled anti-MMR Nb resulted in a significantly retarded tumor growth, thereby outcompeting the effects of anti-PD1, anti-VEGFR2, doxorubicin and paclitaxel in the TS/A mammary carcinoma model. Together, these data propose anti-MMR STRIT as a valid new approach for cancer treatment.


Asunto(s)
Adenocarcinoma/radioterapia , Neoplasias Mamarias Experimentales/radioterapia , Radioinmunoterapia/métodos , Anticuerpos de Dominio Único/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Progresión de la Enfermedad , Doxorrubicina/farmacología , Femenino , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos BALB C , Paclitaxel/farmacología , Receptores de Superficie Celular/metabolismo , Células del Estroma/inmunología
12.
PLoS One ; 14(9): e0222022, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31479484

RESUMEN

In the United States, breast cancer is one of the most common and the second leading cause of cancer-related death in women. Treatment modalities for mammary tumor are surgical removal of the tumor tissue followed by either chemotherapy or radiotherapy or both. Radiation therapy is a whole body irradiation regimen that suppresses the immune system leaving hosts susceptible to infection or secondary tumors. Boron neutron capture therapy (BNCT) in that regard is more selective, the cells that are mostly affected are those that are loaded with 109 or more 10B atoms. Previously, we have described that liposomal encapsulation of boron-rich compounds such as TAC and MAC deliver a high payload to the tumor tissue when injected intravenously. Here we report that liposome-mediated boron delivery to the tumor is inversely proportional to the size of the murine mammary (EMT-6) tumors. The plausible reason for the inverse ratio of boron and EMT-6 tumor size is the necrosis in these tumors, which is more prominent in the large tumors. The large tumors also have receding blood vessels contributing further to poor boron delivery to these tumors. We next report that the presence of boron in blood is essential for the effects of BNCT on EMT-6 tumor inhibition as direct injection of boron-rich liposomes did not provide any added advantage in inhibition of EMT-6 tumor in BALB/c mice following irradiation despite having a significantly higher amount of boron in the tumor tissue. BNCT reaction in PBMCs resulted in the modification of these cells to anti-tumor phenotype. In this study, we report the immunomodulatory effects of BNCT when boron-rich compounds are delivered systemically.


Asunto(s)
Terapia por Captura de Neutrón de Boro/métodos , Inmunomodulación/efectos de la radiación , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/radioterapia , Animales , Boro/administración & dosificación , Boro/sangre , Boro/farmacocinética , Línea Celular Tumoral , Citocinas/metabolismo , Femenino , Humanos , Isótopos/administración & dosificación , Isótopos/sangre , Isótopos/farmacocinética , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/efectos de la radiación , Liposomas , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos BALB C , Necrosis , Distribución Tisular
13.
Phys Med Biol ; 64(19): 195007, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31082811

RESUMEN

Multifunction bismuth-based nanoparticles with the ability to display diagnostic and therapeutic functions have drawn extensive attention as theranostic agents in radiation therapy and imaging due to their high atomic number, low toxicity, and low cost. Herein, we tried to introduce multifunction bismuth gadolinium oxide nanoparticles (BiGdO3) as a new theranostic agent for radiation therapy, computed tomography (CT) and magnetic resonance imaging (MRI). After synthesis of BiGdO3 nanoparticles and surface modifications of them with PEG, biocompatibility of the nanoparticles was evaluated by a CCK-8 assay. We investigated dose amplification properties of the nanoparticles using gel dosimetry and in vitro and in vivo assays. According to clonogenic assay radiation, a sensitizer enhancement ratio (SER) of 1.75 and 1.66 (100 µg ml-1-nanoparticles), for MCF-7 and 4T1 cell lines at low energy x-ray was achieved, respectively. Radiation dose enhancement effect of the nanoparticles was proven for high concentrations (500 µg ml-1) by gel dosimetry. For further investigation, in vivo cancer radiotherapy was carried out using female BALB/c mice with 4T1 breast tumors. In vivo results emphasized the radiosensitizing effect of BiGdO3-PEG nanoparticles. Both bismuth and gadolinium provide CT contrast, while gadolinium can be employed for MRI T1 contrast, so we evaluated contrast enhancement of BiGdO3-PEG nanoparticles as a dual-modal imaging agent in MR and CT imaging. Collectively, our experimental results clearly display properties of BiGdO3-PEG nanoparticles as multimodal imaging and radiosensitizing agents. The results show that the nanoparticles deserve further study as a new theranostic agent.


Asunto(s)
Bismuto/farmacología , Medios de Contraste/farmacología , Gadolinio/farmacología , Nanopartículas Multifuncionales , Fármacos Sensibilizantes a Radiaciones/farmacología , Nanomedicina Teranóstica , Animales , Línea Celular Tumoral , Femenino , Humanos , Células MCF-7 , Imagen por Resonancia Magnética , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Neoplasias Mamarias Experimentales/radioterapia , Ratones , Ratones Endogámicos BALB C , Imagen Multimodal , Trasplante de Neoplasias , Radiometría , Radioterapia , Tomografía Computarizada por Rayos X
14.
Radiother Oncol ; 133: 93-99, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30935588

RESUMEN

BACKGROUND: Synchrotron microbeam radiation therapy (MRT) is a new, evolving form of radiotherapy that has potential for clinical application. Several studies have shown in preclinical models that synchrotron MRT achieves equivalent tumor control to conventional radiotherapy (CRT) but with significantly reduced normal tissue damage. METHODS: To explore differences between these two modalities, we assessed the immune cell infiltrate into EMT6.5 mammary tumors after CRT and MRT. RESULTS: CRT induced marked increases in tumor-associated macrophages and neutrophils while there were no increases in these populations following MRT. In contrast, there were higher numbers of T cells in the MRT treated tumors. There were also increased levels of CCL2 by immunohistochemistry in tumors subjected to CRT, but not to MRT. Conversely, we found that MRT induced higher levels of pro-inflammatory genes in tumors than CRT. CONCLUSION: Our data are the first to demonstrate substantial differences in macrophage, neutrophil and T cell numbers in tumors following MRT versus CRT, providing support for the concept that MRT evokes a different immunomodulatory response in tumors compared to CRT.


Asunto(s)
Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/radioterapia , Animales , Línea Celular Tumoral , Quimiocina CCL2/inmunología , Quimiocina CCL2/metabolismo , Femenino , Macrófagos/inmunología , Macrófagos/efectos de la radiación , Ratones , Ratones Endogámicos BALB C , Neutrófilos/inmunología , Neutrófilos/efectos de la radiación , Radioterapia/instrumentación , Radioterapia/métodos , Sincrotrones , Linfocitos T/inmunología , Linfocitos T/efectos de la radiación
15.
Front Immunol ; 10: 317, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30873170

RESUMEN

A systemic immune related response (SIME) of radiotherapy has been occasionally observed on metastatic tumors, but the clinical outcomes remain poor. Novel treatment approaches are therefore needed to improve SIME ratio. We used a combination of hypo-fractionated radiation therapy (H-RT) with low-dose total body irradiation (L-TBI) in a syngeneic mouse model of breast and colon carcinoma. The combination therapy of H-RT and L-TBI potentially enhanced SIME by infiltration of CD8+ T cell and altering the immunosuppressive microenvironment in non-irradiated subcutaneous tumor lesions. The frequency of IFN-γ, as a tumor-specific CD8+ T cells producing, significantly inhibited the secondary tumor growth of breast and colon. Our findings suggest that L-TBI could serve as a potential therapeutic agent for metastatic breast and colon cancer and, together with H-RT, their therapeutic potential is enhanced significantly.


Asunto(s)
Neoplasias del Colon/inmunología , Neoplasias del Colon/radioterapia , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/radioterapia , Hipofraccionamiento de la Dosis de Radiación , Irradiación Corporal Total , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Línea Celular Tumoral , Femenino , Interferón gamma/inmunología , Ratones Endogámicos BALB C , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación
16.
Radiat Prot Dosimetry ; 183(1-2): 126-130, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30535025

RESUMEN

When early radiation damage to biological systems is studied based on the formation of foci at the location of DNA double-strand breaks, the foci observed in irradiated cells either may be induced by ionizing radiation (IR) interactions or they may be due to other causes that lead to observation of foci also in unirradiated cells. Generally, to take account of the latter, additional samples are taken where the exposure to IR is skipped in the protocol. The data analysis relies on statistical independence of the frequency distributions of background and radiation-induced foci. In microscopy, however, the observed spatial patterns of foci are 2D projections of the spatial distributions of foci in the observed cell nuclei. This may lead to missing foci when scoring their number, particularly if projections of foci overlap or coincide. This paper investigates to what extent the statistical independence of the frequency distribution of the number of foci coming from IR interaction or other causes is compromised by foci overlapping.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de la radiación , Neoplasias Mamarias Experimentales/radioterapia , Modelos Estadísticos , Células Tumorales Cultivadas/efectos de la radiación , Animales , Inmunohistoquímica , Ratones , Probabilidad , Radiación Ionizante
17.
Radiat Prot Dosimetry ; 183(1-2): 121-125, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30520984

RESUMEN

An approach based on track-structure calculations has been developed to take account of artefacts occurring during γ-H2AX foci detection in 2D images of samples analyzed through immunocytochemistry. The need of this works stems from the observed saturation in foci yields measured after X-ray doses higher than few grays, hindering an unambiguous quantification of DNA damage and of radiation effectiveness. The proposed modelling approach allows to simulate the observer's point of view for foci scoring, mimicking the selection of a slice Δz of the cell nucleus due to the microscope depth of field, and applying a clustering algorithm to group together damages within a resolution parameter r. Calculation results were benchmarked with experimental measurements at an early time-point for mouse breast cancer cells, irradiated with X-ray doses in the range 0-5 Gy. The model is able to reproduce the saturation in experimental data.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de la radiación , Histonas/efectos de la radiación , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Neoplasias Mamarias Experimentales/radioterapia , Células Tumorales Cultivadas/efectos de la radiación , Algoritmos , Animales , Inmunohistoquímica , Transferencia Lineal de Energía , Ratones , Método de Montecarlo , Efectividad Biológica Relativa , Programas Informáticos , Rayos X
18.
Nat Commun ; 9(1): 2783, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30018330

RESUMEN

The importance of the tumor-associated stroma in cancer progression is clear. However, it remains uncertain whether early events in the stroma are capable of initiating breast tumorigenesis. Here, we show that in the mammary glands of non-tumor bearing mice, stromal-specific phosphatase and tensin homolog (Pten) deletion invokes radiation-induced genomic instability in neighboring epithelium. In these animals, a single dose of whole-body radiation causes focal mammary lobuloalveolar hyperplasia through paracrine epidermal growth factor receptor (EGFR) activation, and EGFR inhibition abrogates these cellular changes. By analyzing human tissue, we discover that stromal PTEN is lost in a subset of normal breast samples obtained from reduction mammoplasty, and is predictive of recurrence in breast cancer patients. Combined, these data indicate that diagnostic or therapeutic chest radiation may predispose patients with decreased stromal PTEN expression to secondary breast cancer, and that prophylactic EGFR inhibition may reduce this risk.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Experimentales/genética , Fosfohidrolasa PTEN/genética , Tolerancia a Radiación/genética , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/radioterapia , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Transformación Celular Neoplásica , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/efectos de la radiación , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Rayos gamma/efectos adversos , Inestabilidad Genómica/efectos de los fármacos , Inestabilidad Genómica/efectos de la radiación , Humanos , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/efectos de la radiación , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/efectos de la radiación , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/radioterapia , Ratones , Fosfohidrolasa PTEN/deficiencia , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/efectos de la radiación
19.
Mol Pharm ; 15(7): 2606-2613, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29787283

RESUMEN

Angiogenesis is widely recognized as one of the hallmarks of cancer. Therefore, imaging and therapeutic agents targeted to angiogenic vessels may be widely applicable in many types of cancer. To this end, the theranostic isotope pair, 86Y and 90Y, were used to create a pair of agents for targeted imaging and therapy of neovasculature in murine breast cancer models using a chimeric anti-CD105 antibody, TRC105. Serial positron emission tomography imaging with 86Y-DTPA-TRC105 demonstrated high uptake in 4T1 tumors, peaking at 9.6 ± 0.3%ID/g, verified through ex vivo studies. Additionally, promising results were obtained in therapeutic studies with 90Y-DTPA-TRC105, wherein significantly ( p < 0.05) decreased tumor volumes were observed for the targeted treatment group over all control groups near the end of the study. Dosimetric extrapolation and tissue histological analysis corroborated trends found in vivo. Overall, this study demonstrated the potential of the pair 86/90Y for theranostics, enabling personalized treatments for cancer.


Asunto(s)
Antineoplásicos/farmacología , Inmunoconjugados/farmacología , Neoplasias Mamarias Experimentales/radioterapia , Neovascularización Patológica/tratamiento farmacológico , Radioinmunoterapia/métodos , Nanomedicina Teranóstica/métodos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Línea Celular Tumoral/trasplante , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/uso terapéutico , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Distribución Tisular , Resultado del Tratamiento , Radioisótopos de Itrio/química , Radioisótopos de Itrio/farmacología , Radioisótopos de Itrio/uso terapéutico
20.
Phys Med Biol ; 63(11): 114001, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29726404

RESUMEN

We investigated the feasibility of using multifunctional Fe3O4/TaO x (core/shell) nanoparticles, developed for use in contrast agents for computed tomography (CT) and magnetic resonance imaging (MRI), as dose-enhancing radiosensitizers. First, to verify the detectability of Fe3O4/TaO x nanoparticles in imaging, in vivo tests were conducted. Approximately 600 mg kg-1 of 19 nm-diameter Fe3O4/TaO x nanoparticles dispersed in phosphate-buffered saline was injected into the tail vein of six Balb/c mice used as tumour (4T1 mammary carcinoma cell) models. Three mice underwent MRI (BioSpec 70/20 USR, Bruker, Billerica, MA, USA) and micro-CT (Inveon, Siemens Preclinical, Knoxville, TN, USA) before and after the injection. The difference between the pre- and post-injection images was quantified by finding the correlation coefficient. The aorta, blood vessel, and liver were clearly seen in the MRI and micro-CT images 60 min after intravenous injection of Fe3O4/TaO x nanoparticles, but the tumour region was not visible in the CT images until after 24 h. There were large differences between the pre- and post-injection images. Second, the therapeutic enhancement dose of nanomaterials was computed via Monte Carlo simulation. Monoenergetic 70- and 150 MeV proton beams irradiated x-ray contrast agent (iodine, BaSO4), MRI contrast agent (gadolinium, Fe3O4), Au, Fe3O4/TaO x (core/shell) nanoparticles and water located at the centre of a 4 × 4 × 4 µm3 water phantom, upon which the dose enhancement ratio (DER) (dose with/without nanoparticles) was computed. When 70 MeV protons irradiated the Au, gadolinium, Fe3O4/TaO x , Fe3O4, iodine, and BaSO4 nanoparticles, the DERs at 1 nm were 15.76, 7.68, 7.82, 6.17, 4.85, and 5.51, respectively. Fe3O4/TaO x nanoparticles have the potential to be used as a multifunctional agent that enhances tumour detection and increases the dose. Dose enhancement with Fe3O4/TaO x was half that with Au. However, Fe3O4/TaO x is much cheaper than Au, and it is expected that tumour targeting combined with magnetic field could overcome the low DER.


Asunto(s)
Neoplasias Mamarias Experimentales/radioterapia , Nanopartículas del Metal/química , Terapia de Protones/métodos , Fármacos Sensibilizantes a Radiaciones/química , Animales , Medios de Contraste/química , Compuestos Férricos/química , Gadolinio/química , Oro/química , Imagen por Resonancia Magnética , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Ratones , Ratones Endogámicos BALB C , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Tantalio/química , Tomografía Computarizada por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...