Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 239
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(15)2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39125647

RESUMEN

This pre-clinical study was designed to demonstrate how vascular disrupting agents (VDAs) should be administered, either alone or when combined with radiation in clinically relevant fractionated radiation schedules, for the optimal anti-tumor effect. CDF1 mice, implanted in the right rear foot with a 200 mm3 murine C3H mammary carcinoma, were injected with various doses of the most potent VDA drug, combretastatin A-1 phosphate (CA1P), under different schedules. Tumors were also locally irradiated with single-dose, or stereotactic (3 × 5-20 Gy) or conventional (30 × 2 Gy) fractionation schedules. Tumor growth and control were the endpoints used. Untreated tumors had a tumor growth time (TGT5; time to grow to 5 times the original treatment volume) of around 6 days. This increased with increasing drug doses (5-100 mg/kg). However, with single-drug treatments, the maximum TGT5 was only 10 days, yet this increased to 19 days when injecting the drug on a weekly basis or as three treatments in one week. CA1P enhanced radiation response regardless of the schedule or interval between the VDA and radiation. There was a dose-dependent increase in radiation response when the combined with a single, stereotactic, or conventional fractionated irradiation, but these enhancements plateaued at around a drug dose of 25 mg/kg. This pre-clinical study demonstrated how VDAs should be combined with clinically applicable fractionated radiation schedules for the optimal anti-tumor effect, thus suggesting the necessary pre-clinical testing required to ultimately establish VDAs in clinical practice.


Asunto(s)
Fraccionamiento de la Dosis de Radiación , Animales , Ratones , Femenino , Estilbenos/farmacología , Estilbenos/administración & dosificación , Ratones Endogámicos C3H , Neovascularización Patológica/radioterapia , Neovascularización Patológica/tratamiento farmacológico , Línea Celular Tumoral , Neoplasias Mamarias Experimentales/radioterapia , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología
2.
J Theor Biol ; 547: 111175, 2022 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-35644483

RESUMEN

PURPOSE: Tumor oxygenation is one of the key features influencing the response of cells to radiation and chemo therapies. This study presents a novel in silico tumor model simulating realistic 3D microvascular structures and related oxygenation maps, featuring regions with different levels and typologies of hypoxia (chronic, acute and anemic). Such model, if integrated into a treatment planning system, could allow evaluations and comparisons of various scenarios when deciding the therapy to administer. METHODS AND MATERIALS: Spherical tumors between 0.6 and 1.5 cm in diameter encompassed uniformly by vascular trees generated starting from pseudo-fractal principles were simulated with a voxel resolution of 10 µm. The approach ensures a continuous transition from a well-perfused rim to a core with poor vascularization. The oxygen diffusion equation in the tumor is solved by a finite difference method. Several quantities, such as the fractal dimension (FD), the microvascular density (MVD) and the hypoxic fraction (HF) were assessed and compared. RESULTS: Different tumors with various degrees of chronic hypoxia were simulated by varying the tumor size and the number of bifurcations in the vascular networks. The simulations showed that for the case of chronically hypoxic tumors, in well-oxygenated volumes FD = 2.53 ± 0.07, MVD = 3460 ± 2180 vessels/mm3 and HF = 4.0 ± 3.4%, while in hypoxic volumes FD = 2.34 ± 0.09, MVD = 365 ± 156 vessels/mm3, HF = 49.8 ± 18.3%. The superimposition of acute or anemic hypoxia accentuated the oxygen deprivation in the core of the volumes. CONCLUSIONS: Tumors varying in diameter and extension of their vasculature were simulated, showing features that define two distinctive subvolumes in terms of oxygenation. The model could be regarded as a testbed for simulations of key radiobiological features governing the tumor response to radio- and chemotherapy and thus for treatment outcome simulations.


Asunto(s)
Neoplasias , Hipoxia de la Célula , Humanos , Hipoxia , Neoplasias/patología , Neovascularización Patológica/radioterapia , Oxígeno
3.
BMC Cancer ; 21(1): 72, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33446132

RESUMEN

BACKGROUND: p-Boronophenylalanine (10BPA) is a powerful 10B drug used in current clinical trials of BNCT. For BNCT to be successful, a high (500 mg/kg) dose of 10BPA must be administered over a few hours. Here, we report BNCT efficacy after rapid, ultralow-dose administration of either tumor vasculature-specific annexin A1-targeting IFLLWQR (IF7)-conjugated 10BPA or borocaptate sodium (10BSH). METHODS: (1) IF7 conjugates of either 10B drugs intravenously injected into MBT2 bladder tumor-bearing mice and biodistribution of 10B in tumors and normal organs analyzed by prompt gamma-ray analysis. (2) Therapeutic effect of IF7-10B drug-mediated BNCT was assessed by either MBT2 bladder tumor bearing C3H/He mice and YTS-1 tumor bearing nude mice. RESULTS: Intravenous injection of IF7C conjugates of either 10B drugs into MBT2 bladder tumor-bearing mice promoted rapid 10B accumulation in tumor and suppressed tumor growth. Moreover, multiple treatments at ultralow (10-20 mg/kg) doses of IF7-10B drug-mediated BNCT significantly suppressed tumor growth in a mouse model of human YTS-1 bladder cancer, with increased Anxa1 expression in tumors and infiltration by CD8-positive lymphocytes. CONCLUSIONS: We conclude that IF7 serves as an efficient 10B delivery vehicle by targeting tumor tissues via the tumor vasculature and could serve as a relevant vehicle for BNCT drugs.


Asunto(s)
Anexina A1/metabolismo , Compuestos de Boro/administración & dosificación , Terapia por Captura de Neutrón de Boro/métodos , Neovascularización Patológica/radioterapia , Fragmentos de Péptidos/metabolismo , Fenilalanina/análogos & derivados , Neoplasias de la Vejiga Urinaria/radioterapia , Animales , Apoptosis , Compuestos de Boro/química , Compuestos de Boro/metabolismo , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Desnudos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Fenilalanina/administración & dosificación , Fenilalanina/química , Fenilalanina/metabolismo , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Exp Dermatol ; 29(7): 659-666, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32434270

RESUMEN

Rosacea is a skin inflammatory condition accompanied by cutaneous signs such as oedema, flushing, erythema, telangiectasia and pustules. Generally, rosacea is triggered by ultraviolet B (UVB) exposure. When exposed to UVB, skin epidermis thickens and produces elevated levels of pro-inflammatory cytokines, especially keratinocyte-related VEGF, a potent angiogenic factor. The upregulations of VEGF expression and its secretion promote the formation of new blood vessels and exacerbates rosacea. In this study, radiofrequency (RF) irradiation reduced keratinocyte proliferation in the epidermal layer, the expressions of pro-inflammatory cytokines, angiogenesis-related inflammatory factors and VEGF in our UVB-induced model of rosacea in vitro and in vivo. RF irradiation attenuated VEGF-induced angiogenesis-associated processes such as tube formation, cell migration and endothelial cell proliferation. Notably, blood vessel densities in the skins of UVB-treated mice and rosacea patients were significantly decreased by RF irradiation. These results provide experimental and molecular evidence regarding the effectiveness of RF irradiation for the treatment of rosacea.


Asunto(s)
Proliferación Celular/efectos de la radiación , Neovascularización Patológica/radioterapia , Terapia por Radiofrecuencia , Rosácea/metabolismo , Rosácea/radioterapia , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Movimiento Celular/efectos de la radiación , Modelos Animales de Enfermedad , Células Endoteliales , Epidermis , Expresión Génica/efectos de la radiación , Humanos , Interleucina-1beta/genética , Queratinocitos , Masculino , Ratones , Neovascularización Patológica/metabolismo , ARN Mensajero/metabolismo , Ondas de Radio , Transducción de Señal/efectos de la radiación , Rayos Ultravioleta , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
5.
Eur J Nucl Med Mol Imaging ; 45(1): 123-131, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28821931

RESUMEN

PURPOSE: Increased angiogenesis is a marker of aggressiveness in many cancers. Targeted radionuclide therapy of these cancers with angiogenesis-targeting agents may curtail this increased blood vessel formation and slow the growth of tumors, both primary and metastatic. CD105, or endoglin, has a primary role in angiogenesis in a number of cancers, making this a widely applicable target for targeted radioimmunotherapy. METHODS: The anti-CD105 antibody, TRC105 (TRACON Pharmaceuticals), was conjugated with DTPA for radiolabeling with 177Lu (t 1/2 6.65 days). Balb/c mice were implanted with 4T1 mammary carcinoma cells, and five study groups were used: 177Lu only, TRC105 only, 177Lu-DTPA-IgG (a nonspecific antibody), 177Lu-DTPA-TRC105 low-dose, and 177Lu-DTPA-TRC105 high-dose. Toxicity of the agent was monitored by body weight measurements and analysis of blood markers. Biodistribution studies of 177Lu-DTPA-TRC105 were also performed at 1 and 7 days after injection. Ex vivo histology studies of various tissues were conducted at 1, 7, and 30 days after injection of high-dose 177Lu-DTPA-TRC105. RESULTS: Biodistribution studies indicated steady uptake of 177Lu-DTPA-TRC105 in 4T1 tumors between 1 and 7 days after injection (14.3 ± 2.3%ID/g and 11.6 ± 6.1%ID/g, respectively; n = 3) and gradual clearance from other organs. Significant inhibition of tumor growth was observed in the high-dose group, with a corresponding significant increase in survival (p < 0.001, all groups). In most study groups (all except the nonspecific IgG group), the body weights of the mice did not decrease by more than 10%, indicating the safety of the injected agents. Serum alanine transaminase levels remained nearly constant indicating no damage to the liver (a primary clearance organ of the agent), and this was confirmed by ex vivo histological analyses. CONCLUSION: 177Lu-DTPA-TRC105, when administered at a sufficient dose, is able to curtail tumor growth and provide a significant survival benefit without off-target toxicity. Thus, this targeted agent could be used in combination with other treatment options to slow tumor growth allowing the other agents to be more effective.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Lutecio/química , Neoplasias Experimentales/radioterapia , Neovascularización Patológica/radioterapia , Radioinmunoterapia/métodos , Radioisótopos/química , Radiofármacos/uso terapéutico , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Endoglina/inmunología , Femenino , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/patología , Ácido Pentético/química , Radiofármacos/efectos adversos , Radiofármacos/síntesis química , Radiofármacos/farmacocinética , Distribución Tisular
6.
Acta Oncol ; 56(11): 1634-1638, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28838284

RESUMEN

INTRODUCTION: Vascular disrupting agents (VDAs) damage tumor vasculature and enhance tumor radiation response. In this pre-clinical study, we combined radiation with the leading VDA in clinical development, combretastatin A-4 phosphate (CA4P), and compared the effects seen in tumors and relevant normal tissues. MATERIAL AND METHODS: Radiation was applied locally to tissues in CDF1 mice to produce full radiation dose-response curves. CA4P (250 mg/kg) was intraperitoneally (i.p.) injected within 30 minutes after irradiating. Response of 200 mm3 foot implanted C3H mammary carcinomas was assessed using percent tumor control at 90 days. Normal tissue effects were evaluated using early responding skin (development of moist desquamation in the foot at 11-30 days), and late responding bladder (50% reduction in reservoir function estimated by cystometry up to 9 months after treatment), and lung (20% increase in ventilation rate measured by plethysmography within 9 months). A Chi-squared test was used for statistical comparisons (significance level of p < .05). RESULTS: The radiation dose controlling 50% of irradiated tumors was 52 Gy. This significantly decreased to 45 Gy with CA4P. The radiation doses inducing a change in skin, bladder and lung response in 50% of mice were 31 Gy, 14 Gy and 12 Gy, respectively. CA4P had no significant effect on the radiation response of any of these normal tissues. CONCLUSIONS: VDAs significantly enhance tumor radiation response, but had absolutely no effect on the radiation response of early or late responding normal tissues.


Asunto(s)
Bibencilos/farmacología , Pulmón/efectos de la radiación , Neoplasias Mamarias Animales/radioterapia , Neovascularización Patológica/radioterapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Piel/efectos de la radiación , Vejiga Urinaria/efectos de la radiación , Animales , Antineoplásicos Fitogénicos/farmacología , Femenino , Rayos gamma , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos C3H , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología
7.
Clin Exp Pharmacol Physiol ; 44(5): 539-548, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28199751

RESUMEN

The purpose of this paper is to assess the relationship between gene polymorphism in angiogenesis-related genes and radiation responses in nasopharyngeal carcinoma (NPC) patients. The genotypes of 180 NPC patients were analyzed by Sequenom MassARRAY. The response evaluation criteria in solid tumours were used for assessing efficacies, and the criteria of the Radiation Therapy Oncology Group or European Organization for Research & Treatment of Cancer were utilized for evaluating acute toxic reactions in response to radiation. Statistical methods included chi-square test, uni- and multivariate logistic regression analyses. Genotypic carriers of rs1800541 GT were at an elevated risk of developing grade 3+ oral mucositis, and a genetic variant of rs5333 was a predictor for a lower occurring risk of grade 2+ radiation-induced xerostomia. EDN1 rs1800541, rs2071942 and rs5370 variants were associated with a significantly higher risk of severe myelosuppression. SNPs in such angiogenesis-related genes as EDN1 rs1800541, rs2071942 & rs5370 and EDNRA rs5333 may serve as useful biomarkers for predicting the outcomes of NPC patients.


Asunto(s)
Carcinoma/genética , Carcinoma/radioterapia , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/radioterapia , Neovascularización Patológica/genética , Neovascularización Patológica/radioterapia , Polimorfismo de Nucleótido Simple/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Endotelina-1/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Carcinoma Nasofaríngeo , Polimorfismo de Nucleótido Simple/efectos de la radiación , Resultado del Tratamiento , Adulto Joven
8.
Nano Lett ; 15(11): 7488-96, 2015 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-26418302

RESUMEN

More than 50% of all cancer patients receive radiation therapy. The clinical delivery of curative radiation dose is strictly restricted by the proximal healthy tissues. We propose a dual-targeting strategy using vessel-targeted-radiosensitizing gold nanoparticles and conformal-image guided radiation therapy to specifically amplify damage in the tumor neoendothelium. The resulting tumor vascular disruption substantially improved the therapeutic outcome and subsidized the radiation/nanoparticle toxicity, extending its utility to intransigent or nonresectable tumors that barely respond to standard therapies.


Asunto(s)
Oro/efectos adversos , Nanopartículas del Metal/efectos adversos , Neoplasias/radioterapia , Neovascularización Patológica/tratamiento farmacológico , Línea Celular Tumoral , Endotelio/efectos de los fármacos , Endotelio/patología , Endotelio/efectos de la radiación , Oro/química , Humanos , Nanopartículas del Metal/administración & dosificación , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neovascularización Patológica/patología , Neovascularización Patológica/radioterapia , Tolerancia a Radiación/efectos de los fármacos , Radioterapia Guiada por Imagen
9.
Int J Urol ; 22(6): 590-5, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25781902

RESUMEN

OBJECTIVES: To examine the effects of combined treatment with sulfoquinovosylacylpropanediol and X-ray irradiation on the remodeling of the prostate cancer microenvironment, including angiogenic and hypoxic characteristics. METHODS: Human prostate cancer cells (DU145 and PC3) were implanted subcutaneously into the right hind legs of athymic nude mice. After the tumor volume reached 100-300mm(3) , 2mg/kg/day sulfoquinovosylacylpropanediol was given intravenously from day0 to day4, and cells were exposed to 4Gy X-ray irradiation on days0 and 3 (for a total of 8Gy). Tumors were fixed and stained for pathological analyses and immunohistochemical evaluations. To analyze vascular normalization, 60mg/kg pimonidazole dissolved in saline was injected intraperitoneally. RESULTS: Combined treatment with sulfoquinovosylacylpropanediol plus X-ray irradiation enhanced growth inhibition in DU145 xenografts. The tumor vessel density in DU145 cells significantly decreased after the combined treatment. Staining for αsmooth muscle actin in vessels was significantly increased. Pimonidazole staining, showing hypoxic lesions, was negative from 72h, but positive at 6 and 24h after the first combined treatment. In contrast, no enhancement of the microenvironment in PC3 xenografts was observed with sulfoquinovosylacylpropanediol plus X-ray irradiation. CONCLUSION: Sulfoquinovosylacylpropanediol could be a novel potent radiosensitizing agent targeting angiogenesis in prostate cancer.


Asunto(s)
Vasos Sanguíneos/efectos de la radiación , Glucolípidos/administración & dosificación , Neovascularización Patológica/radioterapia , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/radioterapia , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Actinas/análisis , Animales , Vasos Sanguíneos/química , Vasos Sanguíneos/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias de la Próstata/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de la radiación
10.
Glycobiology ; 24(10): 921-5, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24973253

RESUMEN

Radiation therapy is a main stay in treating solid tumors and plays a significant role in definitive and adjuvant therapy. Unfortunately, local control remains a challenge, in which the success of radiotherapy is largely dictated by tumor hypoxia, DNA damage repair and the antitumor immune response. Extensive efforts have therefore been devoted to targeting the factors that attenuate tumor radiosensitivity, although with limited success. Mounting evidence suggests that tumor and endothelial cells may utilize galectin-1 (Gal-1) for protection against radiation through several mechanisms. Targeting Gal-1 in combination with radiotherapy provides an exciting approach to address several radiation-prohibitive mechanisms.


Asunto(s)
Galectina 1/metabolismo , Neoplasias/radioterapia , Neovascularización Patológica/radioterapia , Carcinogénesis/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Endoteliales/efectos de la radiación , Galectina 1/química , Humanos , Hipoxia/metabolismo , Hipoxia/patología , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Tolerancia a Radiación/genética
11.
Br J Cancer ; 111(7): 1310-8, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25077442

RESUMEN

BACKGROUND: Cetuximab is often combined with radiotherapy in advanced SCCHN. Alternative routes bypassing inhibition of EGFR with cetuximab may overshadow the efficacy of this combination. We undertook this study to investigate a possible role of dasatinib in this scenario. METHODS: The SCC5, SCC25, SCC29, FaDu and A431 cell lines were assessed in vitro for cell proliferation under cetuximab and dasatinib treatments. In FaDu and A431 cells, dasatinib plus cetuximab resulted in higher proliferation than cetuximab alone. Then, FaDu and A431 cells were implanted into subcutaneous tissue of athymic mice that were irradiated with 30 Gy in 10 fractions over 2 weeks, and treated with cetuximab and dasatinib. Tumour growth, DNA synthesis and angiogenesis were determined. The EGFR, RAS-GTP activity, phosphorylated AKT, ERK1/2, SRC protein levels and VEGF secretion were determined in vitro. RESULTS: The addition of dasatinib to cetuximab and radiotherapy increased tumour growth, DNA synthesis and angiogenesis that were associated with RAS, AKT and ERK1/2 activation, and SRC inhibition in FaDu and A431 cells. CONCLUSIONS: In xenografts derived from these two cell lines, dasatinib did not improve the efficacy of cetuximab combined with radiotherapy. On the contrary, it worsened tumour control achieved by the combination of these two treatments.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neovascularización Patológica/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Cetuximab , Replicación del ADN , Dasatinib , Fraccionamiento de la Dosis de Radiación , Femenino , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/radioterapia , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Tiazoles/administración & dosificación , Carga Tumoral/efectos de los fármacos , Carga Tumoral/efectos de la radiación , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
12.
Tumour Biol ; 35(12): 12205-15, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25164610

RESUMEN

Endoglin is a proliferation-associated cell membrane antigen and overexpressed in the angiogenic vasculature of solid tumors. However, the applications of endoglin (ENG)-targeted radioimmunotheray in hepatocellular carcinoma have not been reported yet. Therefore, the aim of this study was the visualization of both the development of hepatocellular carcinoma (HCC) tumor burden and therapeutic effect with ENG-targeted (131)I-anti-ENG mAb (A8), via in vivo noninvasive fluorescence imaging (NIFLI) of SMMC7721-green fluorescent protein (GFP) cells. A8 showed a dose-dependent, time-dependent suppression on the proliferation of SMMC7721-GFP cells and human umbilical vein endothelial cells (HUVECs) in vitro. Tube formation assay showed that (131)I-A8 markedly inhibits HUVECs to form extensive and enclosed tube networks. The results showed that the radiochemical purity of (131)I-A8 was 92.8 % and (131)I-A8 maintained more stable in serum than in saline and had high affinity against SMMC7721-GFP cells. The pharmacokinetics of (131)I-A8 was in accordance with the two-compartment model, with a rapid distribution phase and a slow decline phase. NIFLI exhibited a good relation between the fluorescent signal and tumor volume in vivo. Furthermore, treatment with (131)I-A8 resulted in significant tumor-growth suppression on the basis of the reducing fluorescent signal and a remarkably decreased tumor weight in treated animals. These results were further verified by RT-PCR and immunohistochemistry staining. Our findings indicate that (131)I-A8 can be used as ENG-targeted therapy for hepatocellular carcinoma, and noninvasive fluorescence imaging provides valuable information on tumor burden and effectiveness of therapy.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/radioterapia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/radioterapia , Radioinmunoterapia , Receptores de Superficie Celular/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/genética , Antígenos CD/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Modelos Animales de Enfermedad , Endoglina , Humanos , Radioisótopos de Yodo/química , Radioisótopos de Yodo/farmacología , Radioisótopos de Yodo/uso terapéutico , Neoplasias Hepáticas/patología , Masculino , Neovascularización Patológica/metabolismo , Neovascularización Patológica/radioterapia , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Carga Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
13.
World J Urol ; 32(1): 59-67, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23652926

RESUMEN

INTRODUCTION: Renal cell carcinoma is counted among the most resistant tumors to chemotherapy and radiotherapy, respectively. However, therapeutic options expanded since the introduction of molecular agents, targeting specific pathways such as the vascular endothelial growth factor (VEGF)-α, the VEGF receptor (VEGFR), or the mammalian target of rapamycin (mTOR) pathway. These new agents almost doubled the time to tumor progression and in some trials even improved overall survival. Against this background, the role of local treatment strategies in metastasized or inoperable primary renal cell carcinoma has to be redefined. With the onset of new technical developments in radiotherapy and the possibility to precisely deliver higher doses per fraction, encouraging response and control rates have been reported for kidney cancer, supporting a possible role for irradiation in this setting. This overview summarizes the preclinical data and clinical experiences of modern radiotherapy with focus on possible synergies and toxicities when combined with molecular targeted agents. METHODS: The available literature on preclinical and clinical data comprising prospective trials, retrospective analyses and case reports was reviewed. CONCLUSION: With the recent developments in stereotactic and image-guided radiotherapy, encouraging data concerning local control in the treatment for metastasized renal cell carcinoma have been generated and are therefore recommended whenever possible. It seems that with these high- precision irradiation schedules, the combination with targeted agents is feasible with no increase in severe adverse events. Nevertheless, the addition of molecular targeted drugs to radiotherapy outside of approved regimens or clinical trials warrants careful consideration for every single case.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células Renales/terapia , Neoplasias Renales/terapia , Terapia Molecular Dirigida , Radioterapia , Terapia Combinada , Relación Dosis-Respuesta en la Radiación , Humanos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/radioterapia , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
14.
Future Oncol ; 10(15): 2359-79, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25525845

RESUMEN

Radiation therapy can successfully ablate tumors. However, the same ionization process that destroys a cancer can also permanently damage surrounding organs resulting in unwanted clinical morbidity. Therefore, modern radiation therapy attempts to minimize dose to normal tissue to prevent side effects. Still, as tumors and normal tissues intercalate, the risk of normal tissue injury often may prevent tumoricidal doses of radiation therapy to be delivered. This paper will review current outcomes and limitations of radiobiological modifiers that may selectively enhance the radiosensitivity of tumors as well as parallel techniques that may protect normal tissues from radiation injury. Future endeavors based in part upon newly elucidated genetic pathways will be highlighted.


Asunto(s)
Neoplasias/radioterapia , Traumatismos por Radiación/prevención & control , Protectores contra Radiación/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Humanos , Neoplasias/irrigación sanguínea , Neovascularización Patológica/radioterapia , Oncología por Radiación , Tolerancia a Radiación
15.
Future Oncol ; 10(15): 2381-90, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25525846

RESUMEN

Angiotensin II receptor antagonists inhibit various signaling pathways involved in the regulation of inflammation, apoptosis and angiogenesis. Radiation-induced activation of a proinflammatory cytokine network has been shown to mediate normal tissue injury induced by ionizing radiation in cancer patients, resulting in serious side effects. Hence, not only do angiotensin II receptor antagonists block inflammatory signaling both in cancer cells and in normal cells, but they are also effective in the treatment of cancer by inhibiting tumor progression, vascularization and metastasis. This review addresses the role of angiotensin II inhibitors in cancer therapy, and their potential to increase therapeutical index by protecting normal cells and sensitizing tumor cells to radiotherapy.


Asunto(s)
Bloqueadores del Receptor Tipo 2 de Angiotensina II/uso terapéutico , Neoplasias/radioterapia , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Animales , Quimioradioterapia , Humanos , Neovascularización Patológica/radioterapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Transducción de Señal , Resultado del Tratamiento
16.
J Therm Biol ; 44: 55-62, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25086974

RESUMEN

Laser-induced hyperthermia treatment of tumor in a 2-D axisymmetric tissue embedded with moderate size (100-150µm) blood vessels is studied. Laser absorption is enhanced by embedding gold-silica nanoshells in the tumor. Heat transfer in the tissue is modeled using Weinbaum-Jiji bioheat transfer equation. With laser irradiation, the volumetric radiation is accounted in the governing bioheat equation. Radiative information needed in the bioheat equation is calculated using the discrete ordinate method, and the coupled bioheat-radiation equation is solved using the finite volume method. Effects of power density, laser exposure time, beam radius, diameter of blood vessel and volume fractions of nanoshells on temperature spread in the tissue are analyzed.


Asunto(s)
Terapia por Luz de Baja Intensidad , Modelos Biológicos , Neovascularización Patológica/radioterapia , Animales , Oro/química , Humanos , Nanocáscaras/química
17.
Indian J Biochem Biophys ; 51(6): 527-30, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25823226

RESUMEN

Ionizing radiation is a non-specific, but most widely used therapeutic method for cancer treatment. However, a minor fraction of tumor cell population manages to survive after radiation. Radiation efficacy depends on adequate oxygen supply. Rapid growing tumors cause hypoxia that upregulates many pro-survival pathways. At clinical doses, radiation activates inflammatory pathways and causes oxidative stress that plays a positive role during angiogenesis. Selective targeting of signaling mechanisms may radiosensitize tumors.


Asunto(s)
Citocinas/inmunología , Neoplasias/inmunología , Neoplasias/radioterapia , Neovascularización Patológica/inmunología , Neovascularización Patológica/radioterapia , Oxígeno/inmunología , Radioterapia Conformacional/métodos , Animales , Hipoxia de la Célula/inmunología , Hipoxia de la Célula/efectos de la radiación , Proliferación Celular/efectos de la radiación , Humanos , Modelos Inmunológicos , Radiación Ionizante , Resultado del Tratamiento
18.
Endokrynol Pol ; 75(4): 412-418, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39279310

RESUMEN

BACKGROUND: Neuroendocrine tumours (NETs) are a heterogeneous group of tumours, which is characterized by rich vascularization. The role of angiogenesis in NETs has been widely researched. Peptide receptor radionuclide therapy (PRRT) is an effective treatment method for patients with disease progression in NETs. Due to the heterogeneousness of NETs, the response to treatment varies. Currently, the finding of efficient markers helpful in assessing the response to treatment in NETs is crucial. The aim of this study was to assess chromogranin A (CgA) and angiogenic factors in gastro-entero-pancreatic (GEP) and broncho-pulmonary (BP) NET patients treated with PRRT. MATERIAL AND METHODS: The study group included 40 patients with GEP NETs and BP NETs who completed four cycles of PRRT. Serum levels of CgA and angiogenic factors such as vascular endothelial growth factor (VEGF), its receptors (VEGF-R1, VEGF-R2, VEGF-R3), were assessed before and after four cycles of PRRT. All tests were determined using ELISA. RESULTS: The concentration of CgA, VEGF-R1 and VEGF-R2 decreased significantly, whereas VEGF-R3 increased significantly after PRRT. PRRT did not affect VEGF, it was similar before and after the radioisotope treatment. Based on AUROC, only for VEGF-R1 AUC was a consequence of 0.7 which can be considered as a good response to PRRT treatment. CONCLUSIONS: VEGF-R1 may be a potential biomarker useful in assessing the effectiveness of PRRT in NET patients.


Asunto(s)
Cromogranina A , Tumores Neuroendocrinos , Humanos , Tumores Neuroendocrinos/radioterapia , Tumores Neuroendocrinos/sangre , Femenino , Masculino , Persona de Mediana Edad , Adulto , Anciano , Cromogranina A/sangre , Receptores de Péptidos/metabolismo , Biomarcadores de Tumor/sangre , Factor A de Crecimiento Endotelial Vascular/sangre , Neovascularización Patológica/radioterapia , Neovascularización Patológica/sangre , Octreótido/análogos & derivados , Octreótido/uso terapéutico , Neoplasias Pancreáticas/radioterapia , Neoplasias Pancreáticas/sangre , Resultado del Tratamiento
19.
PLoS One ; 19(7): e0304670, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38968211

RESUMEN

In gold nanoparticle-enhanced radiotherapy, intravenously administered nanoparticles tend to accumulate in the tumor tissue by means of the so-called permeability and retention effect and upon irradiation with x-rays, the nanoparticles release a secondary electron field that increases the absorbed dose that would otherwise be obtained from the interaction of the x-rays with tissue alone. The concentration of the nanoparticles in the tumor, number of nanoparticles per unit of mass, which determines the total absorbed dose imparted, can be measured via magnetic resonance or computed tomography images, usually with a resolution of several millimeters. Using a tumor vasculature model with a resolution of 500 nm, we show that for a given concentration of nanoparticles, the dose enhancement that occurs upon irradiation with x-rays greatly depends on whether the nanoparticles are confined to the tumor vasculature or have already extravasated into the surrounding tumor tissue. We show that, compared to the reference irradiation with no nanoparticles present in the tumor model, irradiation with the nanoparticles confined to the tumor vasculature, either in the bloodstream or attached to the inner blood vessel walls, results in a two to three-fold increase in the absorbed dose to the whole tumor model, with respect to an irradiation when the nanoparticles have already extravasated into the tumor tissue. Therefore, it is not enough to measure the concentration of the nanoparticles in a tumor, but the location of the nanoparticles within each volume element of a tumor, be it inside the vasculature or the tumor tissue, needs to be determined as well if an accurate estimation of the resultant absorbed dose distribution, a key element in the success of a radiotherapy treatment, is to be made.


Asunto(s)
Oro , Nanopartículas del Metal , Oro/química , Nanopartículas del Metal/química , Animales , Ratones , Neoplasias/radioterapia , Neoplasias/diagnóstico por imagen , Neoplasias/irrigación sanguínea , Humanos , Dosificación Radioterapéutica , Neovascularización Patológica/radioterapia , Neovascularización Patológica/diagnóstico por imagen
20.
Int J Radiat Biol ; 100(10): 1416-1425, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39101819

RESUMEN

BACKGROUND: Ultrasound-stimulated microbubble (USMB) therapy has proven efficacy of targeting tumor vasculature and enhancing the effect of radiation in tumor xenografts. In this investigation, we studied whether this treatment enhances the sensitivity of cervical cancer to radiation. METHODS: Human cervical cancer (ME-180 and SiHa) cells were treated with USMB or exposed to radiation (0, 2, 4, 6 and 8 Gy) or radiation (8 Gy) in combination with USMB. Clone formation assay and CCK-8 assay were used to analyze the proliferation capacity of cells. Apoptosis and DNA double-strand breaks were detected using flow cytometry and immunofluorescence staining of gamma-H2AX (γ-H2AX), respectively. Matrigel tubule formation was performed to evaluate the angiogenesis of human umbilical vein endothelial cells. In xenograft model of SiHa cells, tumor tissue expression of CD31 was detected by immunohistochemistry. RESULTS: USMB and radiation synergistically restrained the growth of ME-180 and SiHa cells. USMB promoted radiation-induced apoptosis by enhancing the levels of proapoptotic proteins. Furthermore, USMB enhanced radiation-induced γ-H2AX foci to induce DNA double-strand breaks in cervical cancer cells. USMB in combination with radiation reduced the angiogenic capacity of endothelial cells in vitro. Moreover, USMB strengthened the inhibitory effect of radiation on tumor growth and angiogenesis in xenograft models. CONCLUSION: In conclusion, USMB exposure effectively enhanced the destructive effect of radiation on cervical cancer, suggesting that USMB might be a promising sensitizer of radiotherapy to treat cervical cancer.


Asunto(s)
Apoptosis , Microburbujas , Tolerancia a Radiación , Neoplasias del Cuello Uterino , Neoplasias del Cuello Uterino/radioterapia , Neoplasias del Cuello Uterino/irrigación sanguínea , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/diagnóstico por imagen , Microburbujas/uso terapéutico , Humanos , Femenino , Animales , Línea Celular Tumoral , Apoptosis/efectos de la radiación , Ratones , Roturas del ADN de Doble Cadena/efectos de la radiación , Células Endoteliales de la Vena Umbilical Humana , Ondas Ultrasónicas , Ratones Desnudos , Terapia por Ultrasonido/métodos , Histonas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Neovascularización Patológica/radioterapia , Proliferación Celular/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA