Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Photochem Photobiol B ; 203: 111737, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31862636

RESUMEN

Metal nanostructures are promising agents sensitizing by laser light and X-ray in photothermal therapy (PTT) and radiotherapy (RT) of cancer that improve treatment strategies of cancer. Nanoscale platinum materials are favorable in nanomedicine applications. In this study, platinum nanoparticles (PtNPs) were synthesized and applied for cancer therapy upon 808-nm laser light and X-ray radiation, or their combination. Two power densities of laser (1.0 and 1.5 W cm-2) and three X-ray doses (2, 4 and 6 Gy) were selected for irradiation of B16/F10 cell line at 24 and 72 h-post treatment. The synthesized PtNPs had a spherical shape with a diameter of 12.2 ± 0.7 nm, and were cytocompatible up to 250 µg mL-1. A photothermal conversion activity in a concentration-dependent manner at 72 h-post treatment was observed. Also, PtNPs represented cytotoxicity upon X-ray radiation doses of 2, 4, and 6 Gy after 24 h, while, 72-h time passing led to deeper outcomes. Dual radiation of laser light and X-ray into PtNPs considerably improved the treatment via reactive oxygen species (ROS) production. PtNPs can act as a novel dual absorber of laser light and X-ray, a common sensitizer, for treatment of cancer. The results of this study can be considered after further clinical investigations for treatment of tumor models.


Asunto(s)
Apoptosis/efectos de los fármacos , Rayos Láser , Nanopartículas del Metal/toxicidad , Platino (Metal)/química , Animales , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/radioterapia , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Ratones , Nanomedicina , Fototerapia , Especies Reactivas de Oxígeno/metabolismo , Rayos X
2.
Int J Radiat Oncol Biol Phys ; 100(3): 794-801, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29413289

RESUMEN

PURPOSE: This work investigates a new approach to enhance radiotherapy through a photo therapeutic agent activated by Cherenkov light produced from the megavoltage photon beam. The process is termed Radiotherapy Enhanced with Cherenkov photo-Activation (RECA). RECA is compatible with various photo-therapeutics, but here we focus on use with psoralen, an ultraviolet activated therapeutic with extensive history of application in superficial and extracorporeal settings. RECA has potential to extend the scope of psoralen treatments beyond superficial to deep seated lesions. METHODS AND MATERIALS: In vitro studies in B16 melanoma and 4T1 murine breast cancer cells were performed to investigate the potential of RT plus RECA versus RT alone for increasing cytotoxicity (local control) and increasing surface expression of major histocompatibility complex I (MHC I). The latter represents potential for immune response amplification (increased antigen presentation), which has been observed in other psoralen therapies. Cytotoxicity assays included luminescence and clonogenics. The MHC I assays were performed using flow cytometry. In addition, Cherenkov light intensity measurements were performed to investigate the possibility of increasing the Cherenkov light intensity per unit dose from clinical megavoltage beams, to maximize psoralen activation. RESULTS: Luminescence assays showed that RECA treatment (2 Gy at 6 MV) increased cytotoxicity by up to 20% and 9.5% for 4T1 and B16 cells, respectively, compared with radiation and psoralen alone (ie, Cherenkov light was blocked). Similarly, flow cytometry revealed median MHC I expression was significantly higher in RECA-treated cells, compared with those receiving radiation and psoralen alone (approximately 450% and 250% at 3 Gy and 6 Gy, respectively, P << .0001). Clonogenic assays of B16 cells at doses of 6 Gy and 12 Gy showed decreases in tumor cell viability of 7% (P = .017) and 36% (P = .006), respectively, when Cherenkov was present. CONCLUSION: This work demonstrates for the first time the potential for photo-activation of psoralen directly in situ, from Cherenkov light generated by a clinical megavoltage treatment beam.


Asunto(s)
Ficusina/uso terapéutico , Complejo Mayor de Histocompatibilidad , Neoplasias Mamarias Animales/radioterapia , Melanoma Experimental/radioterapia , Fármacos Fotosensibilizantes/uso terapéutico , Fototerapia/métodos , Animales , Supervivencia Celular , Estudios de Factibilidad , Femenino , Mediciones Luminiscentes/métodos , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Ratones , Radioterapia/métodos
3.
Cell Death Dis ; 6: e1761, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25973681

RESUMEN

One prerequisite that radiotherapy (RT) and chemotherapy (CT) result in anti-tumor immune responses is triggering of immunogenic cell death forms such as necroptosis. The latter is inducible by inhibition of apoptosis with the pan-caspase inhibitor zVAD-fmk. The design of multimodal therapies that overcome melanoma's resistance to apoptosis is a big challenge of oncoimmunology. As hints exist that immune stimulation by hyperthermia (HT) augments the efficacy of melanoma therapies and that tumors can be sensitized for RT with zVAD-fmk, we asked whether combinations of RT with dacarbazine (DTIC) and/or HT induce immunogenic melanoma cell death and how this is especially influenced by zVAD-fmk. Necroptosis was inducible in poorly immunogenic B16-F10 melanoma cells and zVAD-fmk generally increased melanoma cell necrosis concomitantly with the release of HMGB1. Supernatants (SNs) of melanoma cells whose cell death was modulated with zVAD-fmk induced an upregulation of the activation markers CD86 and MHCII on macrophages. The same was seen on dendritic cells (DCs), but only when zVAD-fmk was added to multimodal tumor treatments including DTIC. DCs of MyD88 KO mice and DCs incubated with SNs containing apyrase did not increase the expression of these activation markers on their surface. The in vivo experiments revealed that zVAD-fmk decreases the tumor growth significantly and results in a significantly reduced tumor infiltration of Tregs when added to multimodal treatment of the tumor with RT, DTIC and HT. Further, a significantly increased DC and CD8+ T-cell infiltration into the tumor and in the draining lymph nodes was induced, as well as an increased expression of IFNγ by CD8+ T cells. However, zVAD-fmk did not further reduce tumor growth in MyD88 KO mice, mice treated with apyrase or RAG KO mice. We conclude that HMGB1, nucleotides and CD8+ T cells mediate zVAD-fmk induced anti-melanoma immune reactions in multimodal therapy settings.


Asunto(s)
Clorometilcetonas de Aminoácidos/uso terapéutico , Apoptosis/inmunología , Linfocitos T CD8-positivos/inmunología , Proteína HMGB1/metabolismo , Melanoma Experimental/patología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Apirasa/uso terapéutico , Antígeno B7-2/biosíntesis , Antígeno B7-2/inmunología , Inhibidores de Caspasas/uso terapéutico , Línea Celular Tumoral , Quimioradioterapia , Terapia Combinada , Dacarbazina/uso terapéutico , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase II/biosíntesis , Antígenos de Histocompatibilidad Clase II/inmunología , Proteínas de Homeodominio/genética , Hipertermia Inducida , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Activación de Linfocitos/inmunología , Macrófagos Peritoneales/inmunología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/radioterapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Radiación Ionizante , Factor de Necrosis Tumoral alfa/metabolismo
4.
PLoS One ; 10(4): e0124136, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25875081

RESUMEN

Electromagnetic field (EMF) exposures affect many biological systems. The reproducibility of these effects is related to the intensity, duration, frequency, and pattern of the EMF. We have shown that exposure to a specific time-varying EMF can inhibit the growth of malignant cells. Thomas-EMF is a low-intensity, frequency-modulated (25-6 Hz) EMF pattern. Daily, 1 h, exposures to Thomas-EMF inhibited the growth of malignant cell lines including B16-BL6, MDA-MB-231, MCF-7, and HeLa cells but did not affect the growth of non-malignant cells. Thomas-EMF also inhibited B16-BL6 cell proliferation in vivo. B16-BL6 cells implanted in syngeneic C57b mice and exposed daily to Thomas-EMF produced smaller tumours than in sham-treated controls. In vitro studies showed that exposure of malignant cells to Thomas-EMF for > 15 min promoted Ca(2+) influx which could be blocked by inhibitors of voltage-gated T-type Ca(2+) channels. Blocking Ca(2+) uptake also blocked Thomas-EMF-dependent inhibition of cell proliferation. Exposure to Thomas-EMF delayed cell cycle progression and altered cyclin expression consistent with the decrease in cell proliferation. Non-malignant cells did not show any EMF-dependent changes in Ca(2+) influx or cell growth. These data confirm that exposure to a specific EMF pattern can affect cellular processes and that exposure to Thomas-EMF may provide a potential anti-cancer therapy.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Campos Electromagnéticos , Animales , Apoptosis , Calcio/metabolismo , Línea Celular Tumoral , Proliferación Celular , Células HeLa , Humanos , Células MCF-7 , Magnetoterapia , Masculino , Melanoma Experimental/patología , Melanoma Experimental/radioterapia , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Factores de Tiempo , Trasplante Homólogo
5.
J Leukoc Biol ; 97(4): 711-21, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25632046

RESUMEN

RT is commonly used to treat malignant tumors. However, tumor regrowth is a major limitation to RT as an antitumor treatment. In the present study, we investigated the tumor-promoting effects of high-dose (or ablative) RT treatments on tumor-bearing mice. We focused on the role of macrophages that interact with IR-CCs in the TME, which cause tumor regrowth. We observed that CT26(H-2(d)) tumor growth was enhanced by i.v. injection of IR-CT26 cells compared with NR control CT26 cells. The levels of iNOS gene expression and NO production from RAW264.7 macrophages (H-2(d)) in response to the interaction with IR-CT26 cells were higher than with NR-CT26 cells. When CT26 tumor-bearing mice were treated i.v. with L-NMMA, a NOS inhibitor, the reduction in in vivo tumor growth was higher in the IR-CT26-injected group compared with the NR-CT26-injected control group. In vivo CT26 tumor growth was decreased after transplanting PEM extracted from L-NMMA-treated, tumor-bearing mice. Although iNOS activity was reduced by inhibiting TLR1 expression with TLR1-siRNA, it was enhanced by TLR1 overexpression. Transcriptional activation and protein expression levels of iNOS were also decreased in the presence of TLR1-siRNA but increased as a result of TLR1 overexpression. These results demonstrate that postradiotherapeutic tumor regrowth may be caused by interaction of IR-CCs with macrophages that induce TLR1-mediated iNOS expression and NO production. Our data suggest that iNOS in macrophages could be a useful target to regulate postradiotherapeutic responses in hosts and subsequently limit tumor regrowth.


Asunto(s)
Adenocarcinoma/radioterapia , Neoplasias del Colon/radioterapia , Rayos gamma , Macrófagos/metabolismo , Melanoma Experimental/radioterapia , Proteínas de Neoplasias/fisiología , Óxido Nítrico Sintasa de Tipo II/fisiología , Óxido Nítrico/fisiología , Receptor Toll-Like 1/fisiología , Microambiente Tumoral/efectos de la radiación , Células 3T3 , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Células de la Médula Ósea/metabolismo , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Técnicas de Cocultivo , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Progresión de la Enfermedad , Inducción Enzimática , Macrófagos/clasificación , Macrófagos Peritoneales/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Recurrencia , Receptor Toll-Like 1/biosíntesis , Receptor Toll-Like 1/genética , omega-N-Metilarginina/farmacología
6.
Nat Prod Commun ; 9(4): 551-4, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24868882

RESUMEN

The purposes of this study were to investigate the inhibitory effects of Salacia reticulata Tul. root extract on cellular oxidants and melanogenesis in B16 melanoma cells. Cells treated with non-toxic doses of S. reticulata root extract were investigated for their effects on melanogenesis, cellular tyrosinase activity and cellular oxidant scavenging activity. The results indicated that S. reticulata extract inhibited melanin synthesis and tyrosinase activity in alpha-MSH-induced or UV-irradiated B16 melanoma cells in a dose dependent manner. Additionally, the extract also exhibited anti-cellular oxidants in UV-induced radical melanoma cells. Altogether, these results suggested that S. reticulata root extract has roles in suppression of melanogenesis and oxidant inhibition. S. reticulata root extract may be a potential source for the development of pharmaceutical products for treatment of skin hyperpigmentation disorders.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Melaninas/biosíntesis , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/radioterapia , Extractos Vegetales/uso terapéutico , Salacia/química , Animales , Antineoplásicos Fitogénicos/química , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Ratones , Extractos Vegetales/química , Raíces de Plantas/química , Especies Reactivas de Oxígeno/metabolismo
7.
Int J Cancer ; 133(5): 1042-53, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23404099

RESUMEN

The development of alternative therapies for melanoma treatment is of great interest as long-term tumour regression is not achieved with new targeted chemotherapies on selected patients. We previously demonstrated that radioiodinated heteroarylcarboxamide ([131I]ICF01012) induced a strong anti-tumoural effect by inhibiting both primary tumour growth and dissemination process in a B16BL6 melanoma model. In our study, we show that a single injection of [131I]ICF01012 (ranging from 14.8 to 22.2 MBq) was effective and associated with low and transient haematological toxicity. Concerning pigmented organs, cutaneous melanocytes and skin were undamaged. In 30% of treated animals, no histological alteration of retina was observed, and in the remaining 70%, damages were restricted to the optic nerve area. Using the Medical Internal Radiation Dose methodology, we determined that the absorbed dose in major organs is very low (<4 Gy) and that a delivery of 30 Gy to the tumour is sufficient for an effective anti-tumoural response. Molecular analyses of treated tumours showed a strong radiobiological effect with a decrease in proliferation, survival and pro-angiogenic-related markers and an increase in tumour suppressor gene expression, melanogenesis and anti-angiogenic markers. All these features are in accordance with a tumour cell death mechanism that mainly occurs by mitotic catastrophe and provide a better understanding of in vivo anti-tumoural effects of [131I] radionuclide. Our findings raise [131I]ICF01012 a good candidate for disseminated melanoma treatment and strongly support transfer of [131I]ICF01012 to clinical trial.


Asunto(s)
Radioisótopos de Yodo/uso terapéutico , Melaninas/antagonistas & inhibidores , Melanoma Experimental/radioterapia , Quinoxalinas/uso terapéutico , Animales , Ciclo Celular/efectos de la radiación , Humanos , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL
8.
Radiats Biol Radioecol ; 52(5): 510-6, 2012.
Artículo en Ruso | MEDLINE | ID: mdl-23227715

RESUMEN

The elevated efficacy of simultaneous treatment of cancer by irradiation and hyperthermia in comparison with their sequential application was confirmed in the experiments with V-79 cells in vitro and transplantable melanoma B-16 and ELD solid carcinoma in vivo. Irradiation of the tumors transplanted in the calf muscle of mice was performed at the extended Bragg peak of accelerated protons, while extremities of the group of 6 animals were fixed one after another in a water bath at 42 degrees C (hyperthermia) or at 36 degrees C (irradiation at normal temperature). In another series of experiments, the tumors were heated with the microwave (2450 MHz) antennae and irradiated on the 60Co clinical machine. The tumor temperature (42-43 degrees C) was permanently measured with a sensor based on p-i-n diode. In all cases, a hyperthermic treatment lasted 1 h and started either 15 min before irradiation (simultaneous use of both modalities) or 1 h after the end of irradiation, as is usually performed at many institutions. These results, demonstrating a notably better effect of a simultaneous treatment using the cell clonogenicity, the dynamic of tumor regression and the survival of animals as endpoints, made it possible to initiate the development and production of microwave antennae suitable for use during tumor irradiation in clinic.


Asunto(s)
Hipertermia Inducida , Melanoma Experimental/radioterapia , Animales , Línea Celular Tumoral , Cricetinae , Cricetulus , Ratones , Microondas , Protones
9.
Br J Radiol ; 85(1011): 249-58, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22391496

RESUMEN

OBJECTIVES: To evaluate the effects of employing a (10)B-carrier and manipulating intratumour hypoxia on local tumour response and lung metastatic potential in boron neutron capture therapy (BNCT) by measuring the response of intratumour quiescent (Q) cells. METHODS: B16-BL6 melanoma tumour-bearing C57BL/6 mice were continuously given 5-bromo-2'-deoxyuridine (BrdU) to label all proliferating (P) cells. The tumours received reactor thermal neutron beam irradiation following the administration of a (10)B-carrier [L-para-boronophenylalanine-(10)B (BPA) or sodium mercaptoundecahydrododecaborate-(10)B (BSH)] in combination with an acute hypoxia-releasing agent (nicotinamide) or mild temperature hyperthermia (MTH). Immediately after the irradiation, cells from some tumours were isolated and incubated with a cytokinesis blocker. The responses of the Q and total (P+Q) cell populations were assessed based on the frequency of micronuclei using immunofluorescence staining for BrdU. In other tumour-bearing mice, macroscopic lung metastases were enumerated 17 days after irradiation. RESULTS: BPA-BNCT increased the sensitivity of the total tumour cell population more than BSH-BNCT. However, the sensitivity of Q cells treated with BPA was lower than that of BSH-treated Q cells. With or without a (10)B-carrier, MTH enhanced the sensitivity of the Q cell population. Without irradiation, nicotinamide treatment decreased the number of lung metastases. With irradiation, BPA-BNCT, especially in combination with nicotinamide treatment, showed the potential to reduce the number of metastases more than BSH-BNCT. CONCLUSION: BSH-BNCT in combination with MTH improves local tumour control, while BPA-BNCT in combination with nicotinamide may reduce the number of lung metastases.


Asunto(s)
Antineoplásicos/farmacología , Borohidruros/farmacología , Terapia por Captura de Neutrón de Boro/métodos , Hipertermia Inducida/métodos , Melanoma Experimental/radioterapia , Neoplasias Cutáneas/radioterapia , Compuestos de Sulfhidrilo/farmacología , Animales , Bromodesoxiuridina , Hipoxia de la Célula/efectos de los fármacos , Femenino , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Niacinamida/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Complejo Vitamínico B/farmacología
10.
Integr Cancer Ther ; 11(1): 68-80, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21498474

RESUMEN

The present study aimed at evaluating the anticancer and radiosensitizing potential of juglone against a chemoresistant and radioresistant tumor (B16F1 melanoma) growing on C57BL/6J mice. Volume doubling time, growth delay, and median survival were used to assess the in vivo anticancer and radiosensitizing potential of juglone. In vitro radiosensitizing potential of juglone was studied using clonogenic, comet, and reactive oxygen species induction assays. Treatment of tumor-bearing mice with sublethal doses of juglone caused a dose-dependent inhibition of tumor growth as evident from the growth delay and median survival values. Comet assay using tumor tissue and blood showed differential toxicity of juglone, where higher levels of DNA damage was seen in tumor tissue compared with blood cells. Pretreatment of tumor-bearing mice with optimum dose of juglone before radiation resulted in significant tumor growth inhibition compared with radiation alone. From the clonogenic assay, the authors observed a sensitization enhancement ratio of 1.37 for the combination treatment compared with radiation alone. Furthermore, comet assay studies revealed the potential of juglone to enhance the radiation-induced DNA damage and cause a delay in its repair. Juglone pretreatment before radiation also resulted in a significant elevation in the intracellular reactive oxygen species levels compared with radiation alone. In conclusion, the results of this study show the potential of juglone to inhibit the growth of melanoma in vivo. The study also revealed the potential of juglone to augment the radiation-induced cell death of melanoma cells, which may be attributed to oxidative stress-mediated DNA damage and its delayed repair.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Naftoquinonas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Femenino , Masculino , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/radioterapia , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
11.
Cancer Res ; 71(11): 3872-80, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21512134

RESUMEN

Human and rodent solid tumors often exhibit elevated interstitial fluid pressure (IFP). This condition is recognized as a prognostic indicator for reduced responses to therapy and decreased disease-free survival rate. In the present study, we tested whether induction of a thermoregulatory-mediated increase in tissue blood flow, induced by exposure of mice to mild environmental heat stress, could influence IFP and other vascular parameters within tumors. Using several murine tumor models, we found that heating results in a sustained reduction in tumor IFP correlating with increased tumor vascular perfusion (measured by fluorescent imaging of perfused vessels, laser Doppler flowmetry, and MRI) as well as a sustained reduction in tumor hypoxia. Furthermore, when radiation therapy was administered 24 hours postheating, we observed a significant improvement in efficacy that may be a result of the sustained reduction in tumor hypoxia. These data suggest, for the first time, that environmental manipulation of normal vasomotor function is capable of achieving therapeutically beneficial changes in IFP and microvascular function in the tumor microenvironment.


Asunto(s)
Neoplasias del Colon/terapia , Hipertermia Inducida/métodos , Neoplasias Mamarias Experimentales/terapia , Melanoma Experimental/terapia , Animales , Temperatura Corporal/fisiología , Hipoxia de la Célula/fisiología , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/radioterapia , Terapia Combinada , Modelos Animales de Enfermedad , Líquido Extracelular/fisiología , Femenino , Humanos , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/radioterapia , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/radioterapia , Ratones , Ratones Endogámicos BALB C
12.
Integr Cancer Ther ; 9(4): 370-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20713375

RESUMEN

Withaferin A (WA), isolated from Indian medicinal plant Withania somnifera has weak antitumor and radiosensitizing property. The present investigation was planned to evaluate the tumor sensitizing effect of WA with or without local hyperthermia on the response of B16F1 melanoma to fractionated and acute radiotherapy. C57BL mice bearing tumors of 100 ± 10 mm³ were treated with fractionated radiotherapy (RT, 2 Gy x 5 days/week, 4 weeks), withaferin A (15 mg/kg, i.p., 5 days/ week, 3 weeks), local hyperthermia (HT, 43°C once a week, 3 weeks) and their combinations, or acute RT (40 Gy), WA (40 mg/kg), HT (43°C, 30 min) and their combinations. Treatment response was studied by tumor regression, growth delay and animal survival. Acute RT+HT produced 50% partial response which increased to 62.5% with combination of WA. In fractionated regimen, trimodality combination resulted in 100% PR. Acute RT+HT and WA+RT produced similar increase in growth delay (GD) compared to RT alone which further increased in trimodality treatment. Fractionated WA+RT+HT for 3 weeks produced a higher GD and survival than all other treatments. In conclusion, WA is a better radiosensitizer than HT in fractionated regimen and the response of radioresistant tumors like melanoma can be significantly enhanced by combining nontoxic doses of WA with fractionated RT, with or without HT, allowing decrease in radiation dose.


Asunto(s)
Fraccionamiento de la Dosis de Radiación , Hipertermia Inducida , Melanoma Experimental/terapia , Radioterapia Adyuvante/métodos , Neoplasias Cutáneas/terapia , Witanólidos/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Terapia Combinada , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/mortalidad , Melanoma Experimental/radioterapia , Ratones , Ratones Endogámicos C57BL , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/radioterapia , Análisis de Supervivencia , Factores de Tiempo , Resultado del Tratamiento
13.
Br J Radiol ; 83(993): 776-84, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20739345

RESUMEN

The purpose of this study was to evaluate the influence of manipulating intratumour oxygenation status and radiation dose rate on local tumour response and lung metastases following radiotherapy, referring to the response of quiescent cell populations within irradiated tumours. B16-BL6 melanoma tumour-bearing C57BL/6 mice were continuously given 5-bromo-2'-deoxyuridine (BrdU) to label all proliferating (P) cells. They received gamma-ray irradiation at high dose rate (HDR) or reduced dose rate (RDR) following treatment with the acute hypoxia-releasing agent nicotinamide or local hyperthermia at mild temperatures (MTH). Immediately after the irradiation, cells from some tumours were isolated and incubated with a cytokinesis blocker. The responses of the quiescent (Q) and total (proliferating + Q) cell populations were assessed based on the frequency of micronuclei using immunofluorescence staining for BrdU. In other tumour-bearing mice, 17 days after irradiation, macroscopic lung metastases were enumerated. Following HDR irradiation, nicotinamide and MTH enhanced the sensitivity of the total and Q-cell populations, respectively. The decrease in sensitivity at RDR irradiation compared with HDR irradiation was slightly inhibited by MTH, especially in Q cells. Without gamma-ray irradiation, nicotinamide treatment tended to reduce the number of lung metastases. With gamma-rays, in combination with nicotinamide or MTH, especially the former, HDR irradiation decreased the number of metastases more remarkably than RDR irradiation. Manipulating both tumour hypoxia and irradiation dose rate have the potential to influence lung metastasis. The combination with the acute hypoxia-releasing agent nicotinamide may be more promising in HDR than RDR irradiation in terms of reducing the number of lung metastases.


Asunto(s)
Hipoxia de la Célula/efectos de la radiación , Neoplasias Pulmonares/secundario , Melanoma Experimental/radioterapia , Melanoma Experimental/secundario , Niacinamida/administración & dosificación , Animales , Bromodesoxiuridina/administración & dosificación , Hipoxia de la Célula/efectos de los fármacos , Terapia Combinada , Femenino , Rayos gamma/uso terapéutico , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Dosificación Radioterapéutica , Temperatura , Células Tumorales Cultivadas
14.
Cancer Res ; 70(10): 4045-53, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20442292

RESUMEN

Radiolabeled benzamides are attractive candidates for targeted radiotherapy of metastatic melanoma as they bind melanin and exhibit high tumor uptake and retention. One such benzamide, N-(2-diethylamino-ethyl)-4-(4-fluoro-benzamido)-5-iodo-2-methoxy-benzamide (MIP-1145), was evaluated for its ability to distinguish melanin-expressing from amelanotic human melanoma cells, and to specifically localize to melanin-containing tumor xenografts. The binding of [(131)I]MIP-1145 to melanoma cells in vitro was melanin dependent, increased over time, and insensitive to mild acid treatment, indicating that it was retained within cells. Cold carrier MIP-1145 did not reduce the binding, consistent with the high capacity of melanin binding of benzamides. In human melanoma xenografts, [(131)I]MIP-1145 exhibited diffuse tissue distribution and washout from all tissues except melanin-expressing tumors. Tumor uptake of 8.82% injected dose per gram (ID/g) was seen at 4 hours postinjection and remained at 5.91% ID/g at 24 hours, with tumor/blood ratios of 25.2 and 197, respectively. Single photon emission computed tomography imaging was consistent with tissue distribution results. The administration of [(131)I]MIP-1145 at 25 MBq or 2.5 GBq/m(2) in single or multiple doses significantly reduced SK-MEL-3 tumor growth, with multiple doses resulting in tumor regression and a durable response for over 125 days. To estimate human dosimetry, gamma camera imaging and pharmacokinetic analysis was performed in cynomolgus monkeys. The melanin-specific binding of [(131)I]MIP-1145 combined with prolonged tumor retention, the ability to significantly inhibit tumor growth, and acceptable projected human dosimetry suggest that it may be effective as a radiotherapeutic pharmaceutical for treating patients with metastatic malignant melanoma.


Asunto(s)
Benzamidas/uso terapéutico , Radioisótopos de Yodo/uso terapéutico , Melaninas/metabolismo , Melanoma Experimental/radioterapia , Radiofármacos/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Macaca fascicularis , Masculino , Melanoma Experimental/diagnóstico por imagen , Melanoma Experimental/patología , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Dosificación Radioterapéutica , Tasa de Supervivencia , Tomografía Computarizada de Emisión de Fotón Único
15.
J Med Food ; 13(1): 47-53, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20136435

RESUMEN

Although radiotherapy is commonly used for a variety of cancers, radiotherapy alone does not achieve a satisfactory therapeutic outcome. In this study, we examined the possibility that HemoHIM can enhance the anticancer effects of ionizing radiation (IR) in melanoma-bearing mice. The HemoHIM was prepared by adding the ethanol-insoluble fraction to the total water extract of a mixture of three edible herbs-Angelica Radix, Cnidium Rhizoma, and Paeonia Radix. Anticancer effects of HemoHIM were evaluated in melanoma-bearing mice exposed to IR. IR treatment (5 Gy at 7 days after melanoma cell injection) reduced the weight of the solid tumors, and HemoHIM supplementation with IR enhanced the decreases in tumor weight (P < .03). In the melanoma-bearing mice treated with IR, HemoHIM administration also increased the activity of natural killer cells and cytotoxic T cells, although the proportions of these cells in spleen were not different. In addition, HemoHIM administration increased the interleukin-2 and tumor necrosis factor-alpha secretion from lymphocytes stimulated with concanavalin A, which seemed to contribute to the enhanced efficacy of HemoHIM in tumor-bearing mice treated with IR. In conclusion, HemoHIM may be a beneficial supplement during radiotherapy for enhancing the antitumor efficacy.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Magnoliopsida , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/radioterapia , Fitoterapia , Extractos Vegetales/uso terapéutico , Angelica , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Quimioterapia Adyuvante , Cnidium , Concanavalina A , Femenino , Interleucina-2/metabolismo , Células Asesinas Naturales/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitógenos , Paeonia , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacología , Raíces de Plantas , Rizoma , Bazo/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
16.
BMC Cancer ; 9: 404, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19930543

RESUMEN

BACKGROUND: It has been speculated that the biostimulatory effect of Low Level Laser Therapy could cause undesirable enhancement of tumor growth in neoplastic diseases. The aim of the present study is to analyze the behavior of melanoma cells (B16F10) in vitro and the in vivo development of melanoma in mice after laser irradiation. METHODS: We performed a controlled in vitro study on B16F10 melanoma cells to investigate cell viability and cell cycle changes by the Tripan Blue, MTT and cell quest histogram tests at 24, 48 and 72 h post irradiation. The in vivo mouse model (male Balb C, n = 21) of melanoma was used to analyze tumor volume and histological characteristics. Laser irradiation was performed three times (once a day for three consecutive days) with a 660 nm 50 mW CW laser, beam spot size 2 mm(2), irradiance 2.5 W/cm(2) and irradiation times of 60s (dose 150 J/cm(2)) and 420s (dose 1050 J/cm(2)) respectively. RESULTS: There were no statistically significant differences between the in vitro groups, except for an increase in the hypodiploid melanoma cells (8.48 +/- 1.40% and 4.26 +/- 0.60%) at 72 h post-irradiation. This cancer-protective effect was not reproduced in the in vivo experiment where outcome measures for the 150 J/cm(2) dose group were not significantly different from controls. For the 1050 J/cm(2) dose group, there were significant increases in tumor volume, blood vessels and cell abnormalities compared to the other groups. CONCLUSION: LLLT Irradiation should be avoided over melanomas as the combination of high irradiance (2.5 W/cm(2)) and high dose (1050 J/cm(2)) significantly increases melanoma tumor growth in vivo.


Asunto(s)
Proliferación Celular/efectos de la radiación , Terapia por Luz de Baja Intensidad/efectos adversos , Melanoma Experimental/radioterapia , Animales , Apoptosis/efectos de la radiación , Rayos Láser , Masculino , Ratones , Ratones Endogámicos BALB C
17.
Microsc Res Tech ; 72(6): 411-23, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19208388

RESUMEN

We investigated possibilities of the combination of the one- and two-photon excitation microscopy for examination of the experimental melanoma tissue in vivo, in mice under general anesthesia, and ex vivo on freshly harvested specimens. Our aim was to obtain sufficiently informative images of unstained tumor tissues and their modifications after hyperthermia treatment. The mouse experimental melanoma structure was studied and compared with normal tissue from the same animal by using confocal and nonlinear microscopy techniques based on (i) one-photon excitation (1PE) fluorescence, (ii) 1PE reflectance, (iii) second harmonic generation imaging, and (iv) two-photon excitation autofluorescence. We checked different spectral conditions and other settings of image acquisition, as well as combinations of the above imaging modalities, to fully exploit the potential of these techniques in the evaluation of treated and untreated cancer tissue morphology. Our approach enabled to reveal the collagen fiber network in relation with the other tissues, and to identify invasive tumor cells. It also proved to be useful for the examination of interrelationships between functional and morphological aspects based on optical properties of the tissues, especially in studies of changes between the tumor and control tissue, as well as changes induced by physical treatments, e.g., delivery of microwave hyperthermia treatment. These differences were also evaluated quantitatively, when we found out that the maximum Euler-Poincaré characteristic reflects well the melanoma morphological structure. The results showed that the proposed investigative approach could be suitable also for a direct evaluation of tissue modifications induced by clinical interventions.


Asunto(s)
Hipertermia Inducida , Melanoma Experimental/patología , Melanoma Experimental/radioterapia , Microscopía , Animales , Ratones
18.
Int J Hyperthermia ; 17(4): 357-67, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11471986

RESUMEN

PURPOSE: To study the response of a mouse melanoma to radiation and hyperthermia under acute hypoxia and reperfusion. MATERIALS AND METHODS: B16F1 melanoma of 100+/-10 mm3, in C57BL mouse, were locally exposed to 10Gy gamma radiation (RT), 43 degrees C for 30 min (HT) in a water bath, or RT followed immediately by HT, under clamping (acute hypoxia) or 1 h after reperfusion. Tumour regression, volume doubling time (VDT), growth delay (GD), apoptosis and microvascular density (MVD) were studied. RESULTS: Under clamping, HT increased the VDT and GD to > 20 days above control and resulted in > 50% regression (PR) in all the tumours, whilst RT + HT synergistically enhanced VDT and GD. Under reperfusion, HT produced 25% PR against 16% by RT, with no increase in VDT and GD compared to RT. RT + HT significantly enhanced VDT and GD above that of RT or HT, but did not further increase PR of reperfused tumours. HT under clamping caused > 50% increase in apoptic cells over control and decreased MVD to 1/3rd of control. RT + HT further enhanced apoptotic cells to > 70% and reduced MVD to 1/6th of control. CONCLUSIONS: These results suggest that combination of radiotherapy with hyperthermia could benefit treatment of tumours with ischemia-induced acute hypoxia.


Asunto(s)
Hipertermia Inducida , Isquemia , Melanoma Experimental/terapia , Reperfusión , Animales , Apoptosis , División Celular , Terapia Combinada , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Melanoma Experimental/radioterapia , Ratones
19.
Int J Radiat Oncol Biol Phys ; 48(5): 1513-8, 2000 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11121657

RESUMEN

PURPOSE: Interleukin 12 (IL-12) has shown strong antitumoral effects in numerous pre-clinical studies and appears to act synergistically with radiation in murine tumors. The major impediment to its clinical use has been its systemic toxicity. While using intratumorally injected viral gene therapy vectors encoding IL-12 reduces systemic side effects substantially, elevated systemic transgene levels are still observed because adenovirus can reach the circulation. Further restricting IL-12 expression in the tumor is therefore desirable in a combined radiation and adenovirus mediated cancer gene therapy regimen. METHODS AND MATERILAS: Hyperthermia-regulated gene therapy was tested in a nonimmunogenic B16.F10 melanoma line that is syngeneic with C57BL/6 mice. For hyperthermic gene therapy, an adenoviral vector coding for IL-12 under the control of the promoter of the human heat shock protein 70B (hsp70B) was used. One week after transplantation (at a 5-7 mm diameter), tumors were irradiated with 3 x 11 Gy (mo-we-fri). Adenovirus was injected at 3 x 10(8) pfu/tumor 24 h before the last radiation fraction or 3 days afterwards. Hyperthermia was performed 24 h later at 42.5 degrees C. Growth delay to reaching 3 times initial tumor volume was chosen as the biologic endpoint. IL-12 levels in tumor and serum were determined by using the enzyme-linked immunosorbant assay (ELISA). RESULTS: Adenovirus mediated intratumoral expression of IL-12 under the control of a heat inducible promoter in combination with hyperthermia is almost as effective as that under the control of a constitutive cytomegaly virus (CMV) promoter while systemic transgene levels are substantially reduced with the heat inducible promoter. The response to radiotherapy is improved considerably when combined with heat inducible gene therapy without apparent systemic toxicity. When used as a single dose, applying IL-12 gene therapy after completion of radiotherapy appears to be beneficial. CONCLUSIONS: Hyperthermia-regulated gene therapy in combination with radiation is feasible and therapeutically effective in murine tumors with no apparent systemic toxicity.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Hipertermia Inducida , Interleucina-12/uso terapéutico , Melanoma Experimental/terapia , Adenoviridae , Animales , Terapia Combinada , Fraccionamiento de la Dosis de Radiación , Estudios de Factibilidad , Inyecciones Intralesiones , Interleucina-12/genética , Interleucina-12/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/radioterapia , Ratones , Ratones Endogámicos C57BL , Radiobiología
20.
Melanoma Res ; 9(2): 115-24, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10380933

RESUMEN

The purpose of this study was to evaluate the effects of cobalt-60 gamma-radiation and argon laser irradiation using injected merocyanine (MC540) as a photosensitizer on pigmented and non-pigmented Bomirski hamster melanomas growing in the eye. The animals were treated with one of four regimens, receiving gamma-irradiation only, photosensitization only, a combination of gamma-irradiation and photosensitization, or a combined time-fractionated treatment. Tumours were exposed to laser light 24 h after injection, when the photosensitizing dye concentration was highest. The degree of tissue damage was evaluated by observation of the area for necrosis, interruption of blood circulation, and the shape and dissemination of the tumour cells. Additionally, tumour growth was monitored through the measurement of tumour volume and also calculated from histological cross sections on the assumption that the tumour morphology is hemi-ellipsoidal. A single treatment of tumours by a combination of photodynamic therapy and ionizing radiation resulted in an additive effect, inhibiting tumour growth for 2-4 days. A time-fractionated treatment, given four times every 24 h, markedly delayed tumour growth for up to 6 weeks. The results indicate that MC540-mediated photodynamic treatment in combination with gamma-radiation exerts a significant therapeutic effect on a rapidly growing melanoma.


Asunto(s)
Neoplasias del Ojo/radioterapia , Rayos gamma/uso terapéutico , Melanoma Experimental/radioterapia , Trastornos por Fotosensibilidad , Animales , Cricetinae , Relación Dosis-Respuesta en la Radiación , Femenino , Rayos gamma/efectos adversos , Cinética , Mesocricetus , Fototerapia/efectos adversos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA