Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 377
Filtrar
1.
ACS Appl Mater Interfaces ; 13(36): 42473-42485, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34474563

RESUMEN

The particular characteristics of hypoxia, immune suppression in the tumor microenvironment, and the lack of accurate imaging guidance lead to the limited effects of stereotactic body radiotherapy (SBRT) in reducing the recurrence rate and mortality of hepatocellular carcinoma (HCC). This research developed a novel theranostic agent based on Bi/Se nanoparticles (NPs), synthesized by a simple reduction reaction method for in vivo CT image-guided SBRT sensitization in mice. After loading Lenvatinib (Len), the obtained Bi/Se-Len NPs had excellent performance in reversing hypoxia and the immune suppression status of HCC. In vivo CT imaging results uncovered that the radiotherapy (RT) area could be accurately labeled after the injection of Bi/Se-Len NPs. Under Len's unique and robust properties, in vivo treatment was then carried out upon injection of Bi/Se-Len NPs, achieving excellent RT sensitization effects in a mouse HCC model. Comprehensive tests and histological stains revealed that Bi/Se-Len NPs could reshape and normalize tumor blood vessels, reduce the hypoxic situation of the tumor, and upregulate tumor-infiltrating CD4+ and CD8+ T lymphocytes around the tumors. Our work highlights an excellent proposal of Bi/Se-Len NPs as theranostic nanoparticles for image-guided HCC radiotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Medios de Contraste/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Nanopartículas del Metal/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Animales , Bismuto/química , Bismuto/uso terapéutico , Bismuto/toxicidad , Vasos Sanguíneos/efectos de los fármacos , Carcinoma Hepatocelular/diagnóstico por imagen , Línea Celular Tumoral , Medios de Contraste/síntesis química , Medios de Contraste/toxicidad , Portadores de Fármacos/síntesis química , Portadores de Fármacos/uso terapéutico , Portadores de Fármacos/toxicidad , Femenino , Humanos , Hipoxia/tratamiento farmacológico , Neoplasias Hepáticas/diagnóstico por imagen , Linfocitos/efectos de los fármacos , Nanopartículas del Metal/química , Nanopartículas del Metal/toxicidad , Ratones Endogámicos BALB C , Compuestos de Fenilurea/uso terapéutico , Medicina de Precisión , Puntos Cuánticos/química , Puntos Cuánticos/uso terapéutico , Puntos Cuánticos/toxicidad , Quinolinas/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/síntesis química , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/toxicidad , Radiocirugia , Selenio/química , Selenio/uso terapéutico , Selenio/toxicidad , Tomografía Computarizada por Rayos X
2.
ACS Appl Mater Interfaces ; 13(24): 27934-27944, 2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34101408

RESUMEN

Due to conventional photodynamic therapy encountering serious problems of phototoxicity and low tissue-penetrating depth of light, other dynamic therapy-based therapeutic methods such as sonodynamic therapy (SDT) are expected to be developed. To improve the therapeutic response to SDT, more effective sonosensitizers are imperative. In this study, a novel water-soluble iridium(III)-porphyrin sonosensitizer (IrTMPPS) was synthesized and used for SDT. IrTMPPS generated ample singlet oxygen (1O2) under US irradiation and especially showed distinguished US-activatable abilities at more than 10 cm deep-tissue depths. Interestingly, under US irradiation, IrTMPPS sonocatalytically oxidized intracellular NADH, which would enhance SDT efficiency by breaking the redox balance in the tumor. Moreover, IrTMPPS displayed great sonocytotoxicity toward various cancer cells, and in vivo experiments demonstrated efficient tumor inhibition and anti-metastasis to the lungs in the presence of IrTMPPS and US irradiation. This report gives a novel idea of metal-based sonosensitizers for sonotherapy by fully taking advantage of non-invasiveness, water solubility, and deep tumor therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Porfirinas/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/toxicidad , Línea Celular Tumoral , Iridio/química , Iridio/uso terapéutico , Iridio/toxicidad , Ratones , NAD/química , NAD/metabolismo , Neoplasias/patología , Oxidación-Reducción , Porfirinas/síntesis química , Porfirinas/toxicidad , Fármacos Sensibilizantes a Radiaciones/síntesis química , Fármacos Sensibilizantes a Radiaciones/toxicidad , Oxígeno Singlete/metabolismo , Ondas Ultrasónicas , Pez Cebra
3.
Langmuir ; 36(39): 11637-11644, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32902987

RESUMEN

Many noble metal-based nanoparticles have emerged for applications in cancer radiotherapy in recent years, but few investigations have been carried out for palladium nanoparticles. Herein, palladium nanosheets (Pd NSs), which possess a sheetlike morphology with a diameter of ∼14 nm and a thickness of ∼2 nm, were utilized as a sensitizer to improve the performance of radiotherapy. It was found that Pd NSs alone did not decrease the cell viability after treatment for as long as 130 h, suggesting the excellent cytocompatibility of the nanoagents. However, the viability of cancer cells treated with X-ray irradiation became lower, and the viability became even lower if the cells were co-treated with X-ray and Pd NSs, indicating the radiosensitization effect of Pd NSs. Additionally, compared with X-ray irradiation, the combined treatment of Pd NSs and X-ray irradiation induced the generation of more DNA double-stranded breaks and reactive oxygen species within cancer cells, which eventually caused elevated cell apoptosis. Moreover, in vivo experiments also verified the radiosensitization effect and the favorable biocompatibility of Pd NSs, indicating their potential for acquiring satisfactory in vivo radiotherapeutic effect at lower X-ray doses. It is believed that the present research will open new avenues for the application of noble metal-based nanoparticles in radiosensitization.


Asunto(s)
Nanopartículas del Metal , Fármacos Sensibilizantes a Radiaciones , Apoptosis , Supervivencia Celular , Nanopartículas del Metal/toxicidad , Paladio , Fármacos Sensibilizantes a Radiaciones/toxicidad
4.
Biochem Pharmacol ; 182: 114205, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32828802

RESUMEN

A continuous state of oxidative stress during inflammation contributes to the development of 25% of human cancers. Epithelial and inflammatory cells release reactive oxygen species (ROS) and reactive nitrogen species (RNS) that can damage DNA. ROS/RNS have biological implications in both chemoresistance and tumor recurrence. As several clinically employed anticancer drugs can generate ROS/RNS, we have addressed herein how inducible nitric oxide synthase and nitric oxide (iNOS/•NO) affect the molecular pathways implicated in the tumor response to oxidative stress. To mimic the oxidative stress associated with chemotherapy, we used a photosensitizer (pheophorbide a) that can generate ROS/RNS in a controlled manner. We investigated how iNOS/•NO modulates the tumor response to oxidative stress by involving the NF-κB and Nrf2 molecular pathways. We found that low levels of iNOS induce the development of a more aggressive tumor population, leading to survival, recurrence and resistance. By contrast, high levels of iNOS/•NO sensitize tumor cells to oxidative treatment, causing cell growth arrest. Our analysis showed that NF-κB and Nrf2, which are activated in response to oxidative stress, communicate with each other through RKIP. For this critical role, RKIP could be an interesting target for anticancer drugs. Our study provides insight into the complex signaling response of cancer cells to oxidative treatments as well as new possibilities for the rational design of new therapeutic strategies.


Asunto(s)
Óxido Nítrico/fisiología , Estrés Oxidativo/fisiología , Neoplasias de la Próstata/metabolismo , Fármacos Sensibilizantes a Radiaciones/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Clorofila/análogos & derivados , Clorofila/toxicidad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/efectos de la radiación , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo II/efectos de la radiación , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Neoplasias de la Próstata/patología , Especies Reactivas de Oxígeno/efectos de la radiación
5.
Int J Mol Sci ; 21(5)2020 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-32120829

RESUMEN

Nanomedicine has stepped into the spotlight of radiation therapy over the last two decades. Nanoparticles (NPs), especially metallic NPs, can potentiate radiotherapy by specific accumulation into tumors, thus enhancing the efficacy while alleviating the toxicity of radiotherapy. Water radiolysis is a simple, fast and environmentally-friendly method to prepare highly controllable metallic nanoparticles in large scale. In this study, we used this method to prepare biocompatible PEGylated (with Poly(Ethylene Glycol) diamine) platinum nanoflowers (Pt NFs). These nanoagents provide unique surface chemistry, which allows functionalization with various molecules such as fluorescent markers, drugs or radionuclides. The Pt NFs were produced with a controlled aggregation of small Pt subunits through a combination of grafted polymers and radiation-induced polymer cross-linking. Confocal microscopy and fluorescence lifetime imaging microscopy revealed that Pt NFs were localized in the cytoplasm of cervical cancer cells (HeLa) but not in the nucleus. Clonogenic assays revealed that Pt NFs amplify the gamma rays induced killing of HeLa cells with a sensitizing enhancement ratio (SER) of 23%, thus making them promising candidates for future cancer radiation therapy. Furthermore, the efficiency of Pt NFs to induce nanoscopic biomolecular damage by interacting with gamma rays, was evaluated using plasmids as molecular probe. These findings show that the Pt NFs are efficient nano-radio-enhancers. Finally, these NFs could be used to improve not only the performances of radiation therapy treatments but also drug delivery and/or diagnosis when functionalized with various molecules.


Asunto(s)
Nanopartículas del Metal/química , Neoplasias/radioterapia , Platino (Metal)/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Muerte Celular , Citoplasma/metabolismo , Células HeLa , Humanos , Nanopartículas del Metal/toxicidad , Nanopartículas del Metal/ultraestructura , Microscopía Electrónica de Transmisión , Tamaño de la Partícula , Polietilenglicoles/química , Polímeros/química , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/toxicidad , Agua/química
6.
Cancer Prev Res (Phila) ; 13(6): 551-562, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32161072

RESUMEN

Sulforaphane (SFN), a potent antioxidant and antiinflammatory agent, has been shown to protect against cancers especially at early stages. However, how SFN affects UVB-mediated epigenome/DNA methylome and transcriptome changes in skin photodamage has not been fully assessed. Herein, we investigated the transcriptomic and DNA methylomic changes during tumor initiation, promotion, and progression and its impact and reversal by SFN using next-generation sequencing (NGS) technology. The results show that SFN reduced tumor incidence and tumor number. SFN's protective effects were more dramatic in the early stages than with later stages. Bioinformatic analysis of RNA sequencing (RNA-seq) data shows differential expressed genes and identifies the top canonical pathways related to SFN treatment of UVB-induced different stages of epidermal carcinogenesis. These pathways include p53 signaling, cell cycle: G2-M DNA damage checkpoint regulation, Th1, and Th2 activation pathway, and PTEN signaling pathways. The top upstream regulators related to UVB and SFN treatment as time progressed include dextran sulfate, TP53, NFE2L2 (Nrf2), IFNB1, and IL10RA. Bioinformatic analysis of Methyl-seq data shows several differential methylation regions induced by UVB were attenuated by SFN. These include Notch1, Smad6, Gnai3, and Apc2 Integrative analysis of RNA-seq and DNA-seq/CpG methylome yields a subgroup of genes associated with ultraviolet B (UVB) and SFN treatment. The changes in gene expression were inversely correlated with promoter CpG methylation status. These genes include Pik3cd, Matk, and Adm2 In conclusion, our study provides novel insights on the impact of SFN on the transcriptomic and DNA methylomic of UVB-induced different stages of skin cancer in mice.


Asunto(s)
Anticarcinógenos/uso terapéutico , Metilación de ADN/efectos de los fármacos , Epigenoma/efectos de los fármacos , Isotiocianatos/uso terapéutico , Neoplasias Inducidas por Radiación/prevención & control , Neoplasias Cutáneas/prevención & control , Sulfóxidos/uso terapéutico , Transcriptoma/efectos de los fármacos , Rayos Ultravioleta/efectos adversos , Acetona/toxicidad , Animales , Islas de CpG/efectos de los fármacos , ADN de Neoplasias/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Ratones , Ratones Pelados , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neoplasias Inducidas por Radiación/etiología , Neoplasias Inducidas por Radiación/genética , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , RNA-Seq , Fármacos Sensibilizantes a Radiaciones/toxicidad , Distribución Aleatoria , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/genética
7.
J Photochem Photobiol B ; 205: 111820, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32065959

RESUMEN

Recent studies focus on usage of blue light of λ = 450 nm in combination with photosensitizers to treat surface skin disorders, including cancers. In search of convenient therapeutic factor we studied riboflavin analogue 3-methyl-tetraacetylriboflavin (3MeTARF) as potential sensitizer. Riboflavin (Rfl) itself, non -toxic in the darkness, upon absorption of UVA and blue light, may act as photosensitizer. However, Rfl efficiency is limited due to its susceptibility to photodecomposition. Riboflavin's acetylated analogue, 3MeTARF, bears substituents in ribose chain, which inhibit intramolecular processes leading to degradation. Upon excitation, this compound, reveals higher photochemical resistance, remaining a good singlet oxygen generator. Thus, being more stable as the sensitizer, might be much more efficient in photodynamic processes. The objective of undertaken study was to elucidate mechanisms of 3MeTARF photoreactivity under the irradiation with blue light in comparison to its mater compound, riboflavin. We approached this goal by using spectroscopic methods, like direct singlet oxygen phosphorescence detection at 1270 nm, EPR spin trapping and oximetry. Additionally, we tested both riboflavin and 3MeTARF phototoxicity against melanoma cells (WM115) and we studied mechanism of photodynamic cell death, as well. Moreover, 3MeTARF induces apoptosis in melanoma cells at ten times lower concentration than riboflavin itself. Our studies confirmed that 3MeTARF remains stable upon blue light activation and is more efficient photosensitizer than Rfl.


Asunto(s)
Fármacos Sensibilizantes a Radiaciones , Riboflavina , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dermatitis Fototóxica , Humanos , Peróxido de Hidrógeno/metabolismo , Luz , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/efectos de la radiación , Fármacos Sensibilizantes a Radiaciones/toxicidad , Riboflavina/análogos & derivados , Riboflavina/química , Riboflavina/efectos de la radiación , Riboflavina/toxicidad , Oxígeno Singlete/química
8.
Clin Cancer Res ; 25(20): 6035-6043, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31337643

RESUMEN

PURPOSE: Iododeoxyuridine (IUdR) is a potent radiosensitizer; however, its clinical utility is limited by dose-limiting systemic toxicities and the need for prolonged continuous infusion. 5-Iodo-2-pyrimidinone-2'-deoxyribose (IPdR) is an oral prodrug of IUdR that, compared with IUdR, is easier to administer and less toxic, with a more favorable therapeutic index in preclinical studies. Here, we report the clinical and pharmacologic results of a first-in-human phase I dose escalation study of IPdR + concurrent radiation therapy (RT) in patients with advanced metastatic gastrointestinal (GI) cancers. PATIENTS AND METHODS: Adult patients with metastatic GI cancers referred for palliative RT to the chest, abdomen, or pelvis were eligible for study. Patients received IPdR orally once every day × 28 days beginning 7 days before the initiation of RT (37.5 Gy in 2.5 Gy × 15 fractions). A 2-part dose escalation scheme was used, pharmacokinetic studies were performed at multiple time points, and all patients were assessed for toxicity and response to Day 56. RESULTS: Nineteen patients were entered on study. Dose-limiting toxicity was encountered at 1,800 mg every day, and the recommended phase II dose is 1,200 mg every day. Pharmacokinetic analyses demonstrated achievable and sustainable levels of plasma IUdR ≥1 µmol/L (levels previously shown to mediate radiosensitization). Two complete, 3 partial, and 9 stable responses were achieved in target lesions. CONCLUSIONS: Administration of IPdR orally every day × 28 days with RT is feasible and tolerable at doses that produce plasma IUdR levels ≥1 µmol/L. These results support the investigation of IPdR + RT in phase II studies.


Asunto(s)
Quimioradioterapia/métodos , Neoplasias Gastrointestinales/terapia , Idoxuridina/farmacocinética , Nucleósidos de Pirimidina/administración & dosificación , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Fraccionamiento de la Dosis de Radiación , Estudios de Factibilidad , Femenino , Neoplasias Gastrointestinales/patología , Humanos , Idoxuridina/administración & dosificación , Idoxuridina/toxicidad , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Estadificación de Neoplasias , Profármacos/administración & dosificación , Profármacos/farmacocinética , Profármacos/toxicidad , Nucleósidos de Pirimidina/farmacocinética , Nucleósidos de Pirimidina/toxicidad , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Fármacos Sensibilizantes a Radiaciones/toxicidad , Resultado del Tratamiento
9.
Drug Deliv Transl Res ; 9(3): 615-624, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30690675

RESUMEN

Nanocarriers for drug delivery have made great progress in the treatment of cancer, but the dense extracellular collagen of tumors has greatly limited the efficiency of drug delivery. In this study, losartan is used to deplete tumor collagen and improve the delivery efficiency and photodynamic therapeutic efficacy of chlorine 6 (Ce6)-loaded periodic mesoporous organosilica nanoplatform (Ce6-PMO) for breast cancer. After pretreatment with losartan in vivo, the tumor collagen I fraction is significantly reduced by 53% compared to that of mice pretreated with saline. Importantly, the accumulation of the Ce6-PMO nanoplatforms in the tumor is remarkably enhanced via peritumoral and intravenous injection, respectively, after the mice are pretreated with losartan. Further, combination of losartan with the Ce6-PMO nanoplatforms shows the best therapeutic efficacy, and the suppression rate of tumor volume is measured up to 82%. Taken together, this study provides a very promising synergetic strategy to improve the tumor photodynamic treatment efficacy of nanoplatforms.


Asunto(s)
Colágeno Tipo I/metabolismo , Losartán/administración & dosificación , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Fotoquimioterapia , Porfirinas/administración & dosificación , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Dióxido de Silicio/administración & dosificación , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Clorofilidas , Femenino , Losartán/toxicidad , Masculino , Neoplasias Mamarias Experimentales/patología , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Porfirinas/toxicidad , Fármacos Sensibilizantes a Radiaciones/toxicidad , Dióxido de Silicio/toxicidad , Carga Tumoral/efectos de los fármacos
10.
Environ Res ; 170: 383-388, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30623885

RESUMEN

PURPOSE: To evaluate if the common field lampricide 3-trifluoromethyl-4-nitrophenol (TFM) that is intended to eradicate the invasive species sea lampreys in the Great Lakes has the potential to sensitize radiation responses in cells from non-targeted native fish MATERIALS AND METHODS: The TFM toxicity was assessed acutely and chronically with the clonogenic fish cell line eelB. The acute toxicity (24-h exposure) was determined by the fluorescent cell viability probe Alamar Blue. The chronic toxicity was determined either by Alamar Blue (7-d exposure) or the clonogenic survival assay (14-d exposure). Pre- and post-exposure of fish cells to environmentally relevant TFM concentrations following gamma irradiation were performed. Clonogenic survival was determined to assess the damage level of radiation-induced reproductive cell death. RESULTS: The chronic toxicity tests were more sensitive than the acute toxicity tests. The 14-d EC50 using the clonogenic survival endpoint was 2.09 ±â€¯0.28 µg/mL and was statistically similar to the 7-d EC50 (1.85 ±â€¯0.07 µg/mL) based on the Alamar Blue-based cytotoxicity endpoint. Post-exposure of cells to environmentally relevant TFM concentrations following irradiation did not have any effect as compared to the irradiation alone group. In contrast, pre-exposure of cells to TFM following irradiation had a negative additive effect when the total radiation dose was 2 Gy, but not 0.1 or 0.5 Gy. CONCLUSION: Our results suggest that the common field lampricide TFM is a potential radiation sensitizer in cells from non-targeted native fish. This could be a health problem of concern for non-targeted native fish if a large accidental radioactive release occurs.


Asunto(s)
Nitrofenoles/toxicidad , Fármacos Sensibilizantes a Radiaciones/toxicidad , Animales , Supervivencia Celular , Peces , Petromyzon/fisiología
11.
Clin Transl Oncol ; 21(4): 479-488, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30298468

RESUMEN

OBJECTIVES: To investigate the effects of Au@Fe2O3 core-shell nanoparticle (NP), with and without conjugation to folic acid (FA) as a targeting ligand, on radiosensitization of both cancer and healthy cells. METHODS: Au@Fe2O3 NPs were first synthesized, then modified with FA, and finally characterized. Radiation dose enhancement studies were performed on KB cancer cells and L929 healthy cells. NPs at the concentration of 20 µg/ml were first incubated with both cell lines and then different doses of 6 MV X-ray radiation were examined. The end effects were evaluated via MTT assay and flow cytometry using AnnexinV/PI kit. RESULTS: It was indicated that viability of KB cells has a much lower rate than L929 cells when the cells were treated by {(FA-Au@Fe2O3) + (X-ray)} regimen. Cell viability was even decreased significantly when X-ray dose increased. Moreover, flow cytometry studies revealed that FA-targeted NPs induced higher level of apoptosis for KB cancer cells than L929 healthy cells. CONCLUSION: Our findings provide a new perspective on high ability of the synthesized FA-targeted Au@Fe2O3 NPs which may be considered as an efficient radiosensitizer in the process of targeted radiation therapy of cancer.


Asunto(s)
Sistemas de Liberación de Medicamentos , Ácido Fólico/química , Oro/química , Nanopartículas de Magnetita/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Humanos , Células KB , Células L , Ratones , Dosis de Radiación , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/toxicidad , Radioterapia , Rayos X
12.
Biomed Pharmacother ; 109: 2173-2181, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30551474

RESUMEN

Breast cancer resistance protein (BCRP) belongs to the family of ATP-binding cassette (ABC) transporters, overexpression of which can confer a multidrug-resistant phenotype in cancer cells and tumors. BCRP mediates efflux of numerous xenobiotics, including various chemotherapeutic agents and photosensitizers. Hypericin (HY) is a naturally-occurring photosensitizer synthesized by plants of the genus Hypericum. Our recently published results indicate that accumulation of HY in cancer cells of different tissue origin can be affected mostly by BCRP. Considering all known facts, the main goal of this study was to verify whether not only HY accumulation but also toxicity of HY-mediated photodynamic therapy (PDT) can be affected by the presence of some ABC transporters. To specifically prove our hypothesis, we used an experimental model of human leukemia cell lines differing in the expression level of the main drug efflux transporters P-glycoprotein (P-gp), multidrug resistance-associated protein 1 (MRP1) and BCRP. The lowest HY accumulation, and consequently the highest resistance to HY-PDT, was found in cells overexpressing BCRP. Moreover, pretreatment with BCRP inhibitor Ko143 significantly increased HY accumulation and sensitized cells to HY-PDT. Therefore, our findings represent direct evidence that BCRP is the nemesis of HY accumulation and toxicity of HY-PDT. Thus, we should emphasize that individualized screening for BCRP expression and activity may represent a useful tool for prediction of HY-mediated photodynamic diagnosis (PDD) or PDT effectiveness.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Perileno/análogos & derivados , Fotoquimioterapia , Fármacos Sensibilizantes a Radiaciones/metabolismo , Antracenos , Antineoplásicos/metabolismo , Antineoplásicos/toxicidad , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/fisiología , Femenino , Células HL-60 , Humanos , Perileno/antagonistas & inhibidores , Perileno/metabolismo , Perileno/toxicidad , Fotoquimioterapia/efectos adversos , Fármacos Sensibilizantes a Radiaciones/toxicidad
13.
Int J Radiat Biol ; 94(9): 838-843, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29939813

RESUMEN

PURPOSE: Some phytochemicals have shown the potential of being radiomodifiers, especially phenolic compounds, such as lichenic secondary metabolites. To evaluate the phytochemical usnic acid as a radiomodifier, embryonic cells of molluscs have been used due to their ease of collection, high sensitivity to physical and chemical agents, well-known embryology and low cost for analysis. MATERIALS AND METHODS: This study aimed to assess the radiosensitizing action of usnic acid on Biomphalaria glabrata embryos. Samples were irradiated with 4 Gy of gamma rays from a 60Co source (dose rate 2.906 Gy/h). An acute toxicity test was performed using B. glabrata embryos in the blastula stage, in order to determine the toxicity of usnic acid and to establish the lethal Concentration for 50% (LC50). Subsequently, the radiomodifing capacity of usnic acid was estimated using assays with B. glabrata embryos. RESULTS: Irradiation increased the number of non-viable embryos compared to unirradiated controls. Additionally, it was observed that embryos exposed to a non-toxic concentration of usnic acid (0.6 µg/mL) before irradiation showed a further enhancement in non-viable embryos when compared with exposure to ionizing radiation alone. CONCLUSION: The results presented here indicate that usnic acid makes cells more sensitive to the damaging effects of radiation.


Asunto(s)
Benzofuranos/farmacología , Biomphalaria/embriología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/efectos de la radiación , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Benzofuranos/toxicidad , Radioisótopos de Cobalto , Rayos gamma , Dosificación Letal Mediana , Fármacos Sensibilizantes a Radiaciones/toxicidad
14.
J Med Chem ; 61(3): 1241-1254, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29253343

RESUMEN

Innovations in the field of radiotherapy such as stereotactic body radiotherapy, along with the advent of radio-immuno-oncology, herald new opportunities for classical oxygen-mimetic radiosensitizers. The role of hypoxic tumor cells in resistance to radiotherapy and in suppression of immune response continues to endorse tumor hypoxia as a bona fide, yet largely untapped, drug target. Only nimorazole is used clinically as a radiosensitizer, and there is a dearth of new radiosensitizers in development. Here we present a survey of novel nitroimidazole alkylsulfonamides and document their cytotoxicity and ability to radiosensitize anoxic tumor cells in vitro. We use a phosphate prodrug approach to increase aqueous solubility and to improve tumor drug delivery. A 2-nitroimidazole and a 5-nitroimidazole analogue demonstrated marked tumor radiosensitization in either ex vivo assays of surviving clonogens or tumor regrowth delay.


Asunto(s)
Nitroimidazoles/química , Nitroimidazoles/farmacología , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Descubrimiento de Drogas , Femenino , Células HCT116 , Humanos , Ratones , Nitroimidazoles/farmacocinética , Nitroimidazoles/toxicidad , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Fármacos Sensibilizantes a Radiaciones/toxicidad , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
15.
DNA Repair (Amst) ; 60: 89-101, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29112893

RESUMEN

Investigation of natural products is an attractive strategy to identify novel compounds for cancer prevention and treatment. Numerous studies have shown the efficacy and safety of natural products, and they have been widely used as alternative treatments for a wide range of illnesses, including cancers. However, it remains unknown whether natural products affect homologous recombination (HR)-mediated DNA repair and whether these compounds can be used as sensitizers with minimal toxicity to improve patients' responses to radiation therapy, a mainstay of treatment for many human cancers. In this study, in order to systematically identify natural products with an inhibitory effect on HR repair, we developed a high-throughput image-based HR repair screening assay and screened a chemical library containing natural products. Among the most interesting of the candidate compounds identified from the screen was ß-thujaplicin, a bioactive compound isolated from the heart wood of plants in the Cupressaceae family, can significantly inhibit HR repair. We further demonstrated that ß-thujaplicin inhibits HR repair by reducing the recruitment of a key HR repair protein, Rad51, to DNA double-strand breaks. More importantly, our results showed that ß-thujaplicin can radiosensitize cancer cells. Additionally, ß-thujaplicin sensitizes cancer cells to PARP inhibitor in different cancer cell lines. Collectively, our findings for the first time identify natural compound ß-thujaplicin, which has a good biosafety profile, as a novel HR repair inhibitor with great potential to be translated into clinical applications as a sensitizer to DNA-damage-inducing treatment such as radiation and PARP inhibitor. In addition, our study provides proof of the principle that our robust high-throughput functional HR repair assay can be used for a large-scale screening system to identify novel natural products that regulate DNA repair and cellular responses to DNA damage-inducing treatments such as radiation therapy.


Asunto(s)
Monoterpenos/uso terapéutico , Neoplasias/tratamiento farmacológico , Recombinasa Rad51/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Reparación del ADN por Recombinación/efectos de los fármacos , Tropolona/análogos & derivados , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Cupressaceae/química , ADN/metabolismo , Roturas del ADN de Doble Cadena , Quimioterapia Combinada , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Monoterpenos/farmacología , Monoterpenos/toxicidad , Neoplasias/enzimología , Neoplasias/genética , Extractos Vegetales , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Recombinasa Rad51/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Fármacos Sensibilizantes a Radiaciones/toxicidad , Tropolona/farmacología , Tropolona/uso terapéutico , Tropolona/toxicidad
16.
J Coll Physicians Surg Pak ; 27(6): 342-347, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28689522

RESUMEN

OBJECTIVE: To evaluate the efficacy of concurrent chemoradiation in patients with locally advanced inoperable squamous cell carcinoma of oral cavity in terms of local control and toxicity. STUDY DESIGN: Case series. PLACE AND DURATION OF STUDY: Institute of Nuclear Medicine and Oncology (INMOL), Lahore, from January 2008 to December 2013. METHODOLOGY: Sixty-nine patients with locally advanced inoperable oral cavity cancer, registered in INMOL hospital from January 2008 to December 2013 who fulfilled a pre-defined eligibility criteria, were enrolled in the study. Concurrent chemoradiation protocol consisted of conventional fractionation delivering 70 Gy with weekly Cisplatin (50 mg/m2) during the course of radiation. Tumor response was calculated by RECISTcriteria version 1.1 along with the median overall survival and disease-free survival. Acute treatment related toxicities were graded as (G). RESULTS: Thirty-six (52.17%) patients showed complete response; while 19 (27.54%), 8 (11.59%) and 6 (8.7%) were observed with partial response, stable and progressive disease, respectively. Treatment response was significant (p<0.001) in terms of responders vs. non responders to treatment. Median overall survival was 18.00 months; whereas, median disease-free survival remained 14.00 months. Main toxicities included mucositis (G3 and G4, 71%), xerostomia (G2 and G3, 82.5%), vomiting (G3 and G4, 51%), myelosuppression (G3 and G4, 26.2%), dermatitis (G3 and G4, 49.2%), and fatigue (G3 and G4, 57.9%). CONCLUSION: Platinum based CCR Tremained effective for inoperable oral cancer patients.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/radioterapia , Quimioradioterapia/efectos adversos , Cisplatino/administración & dosificación , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/radioterapia , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Adulto , Anciano , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Cisplatino/toxicidad , Terapia Combinada , Fraccionamiento de la Dosis de Radiación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/mortalidad , Neoplasias de la Boca/patología , Mucositis/inducido químicamente , Pakistán , Fármacos Sensibilizantes a Radiaciones/toxicidad , Dosificación Radioterapéutica , Índice de Severidad de la Enfermedad , Tasa de Supervivencia , Resultado del Tratamiento , Vómitos/inducido químicamente , Xerostomía/inducido químicamente
17.
Int J Radiat Biol ; 93(8): 757-763, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28452253

RESUMEN

BACKGROUND AND PURPOSE: The aim of radiotherapy is to deliver lethal damage to cancerous tissue while preserving adjacent normal tissues. Radiation absorbed dose of the tumoral cells can increase when high atomic nanoparticles are present in them during irradiation. Also, the dose rate is an important aspect in radiation effects that determines the biological results of a given dose. This in vitro study investigated the dose-rate effect on the induced radiosensitivity by dextran-coated iron oxide in cancer cells. MATERIALS AND METHODS: HeLa and MCF-7 cells were cultured in vitro and incubated with different concentrations of dextran-coated iron oxide nanoparticles. They were then irradiated with 6 MV photons at dose rates of 43, 185 and 370 cGy/min. The MTT test was used to obtain the cells' survival after 48 h of irradiations. RESULTS: Incubating the cells with the nanoparticles at concentrations of 10, 40 and 80 µg/ml showed no significant cytotoxicity effect. Dextran-coated iron oxide nanoparticles showed more radiosensitivity effect by increasing the dose rate and nanoparticles concentration. Radiosensitization enhancement factors of MCF-7 and HeLa cells at a dose-rate of 370 cGy/min and nanoparticles' concentration of 80 µg/ml were 1.21 ± 0.06 and 1.19 ± 0.04, respectively. CONCLUSION: Increasing the dose rate of 6 MV photons irradiation in MCF-7 and HeLa cells increases the radiosensitization induced by the dextran-coated iron nanoparticles in these cells.


Asunto(s)
Dextranos/química , Compuestos Férricos/química , Compuestos Férricos/farmacología , Nanopartículas , Dosis de Radiación , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Relación Dosis-Respuesta en la Radiación , Compuestos Férricos/toxicidad , Células HeLa , Humanos , Células MCF-7 , Fármacos Sensibilizantes a Radiaciones/toxicidad , Rayos X
18.
Sci Rep ; 6: 31973, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27558808

RESUMEN

Ruthenium(II) polypyridyl complexes can intercalate DNA with high affinity and prevent cell proliferation; however, the direct impact of ruthenium-based intercalation on cellular DNA replication remains unknown. Here we show the multi-intercalator [Ru(dppz)2(PIP)](2+) (dppz = dipyridophenazine, PIP = 2-(phenyl)imidazo[4,5-f][1,10]phenanthroline) immediately stalls replication fork progression in HeLa human cervical cancer cells. In response to this replication blockade, the DNA damage response (DDR) cell signalling network is activated, with checkpoint kinase 1 (Chk1) activation indicating prolonged replication-associated DNA damage, and cell proliferation is inhibited by G1-S cell-cycle arrest. Co-incubation with a Chk1 inhibitor achieves synergistic apoptosis in cancer cells, with a significant increase in phospho(Ser139) histone H2AX (γ-H2AX) levels and foci indicating increased conversion of stalled replication forks to double-strand breaks (DSBs). Normal human epithelial cells remain unaffected by this concurrent treatment. Furthermore, pre-treatment of HeLa cells with [Ru(dppz)2(PIP)](2+) before external beam ionising radiation results in a supra-additive decrease in cell survival accompanied by increased γ-H2AX expression, indicating the compound functions as a radiosensitizer. Together, these results indicate ruthenium-based intercalation can block replication fork progression and demonstrate how these DNA-binding agents may be combined with DDR inhibitors or ionising radiation to achieve more efficient cancer cell killing.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Complejos de Coordinación/química , Replicación del ADN/efectos de los fármacos , Sustancias Intercalantes/química , Fármacos Sensibilizantes a Radiaciones/química , Rutenio/química , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Bovinos , Línea Celular , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Complejos de Coordinación/metabolismo , Complejos de Coordinación/toxicidad , ADN/metabolismo , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de la radiación , Células HeLa , Histonas/metabolismo , Humanos , Sustancias Intercalantes/metabolismo , Sustancias Intercalantes/toxicidad , Fosforilación/efectos de los fármacos , Quinolinas/toxicidad , Quinuclidinas/toxicidad , Radiación Ionizante , Fármacos Sensibilizantes a Radiaciones/metabolismo , Fármacos Sensibilizantes a Radiaciones/toxicidad
19.
J Nanosci Nanotechnol ; 16(5): 4554-60, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27483789

RESUMEN

Adenylate cyclase is a key intracellular enzyme involved in energy imbalance leading to tumor hypoxia and cytotoxicity. In this study, adenylate cyclase activities in isolated hepatocytes and Kupffer cells were compared in the presence of several metabolic stimulators. In cultured hepatocyte cells, adenylate cyclase was stimulated by guanylyl imidotriphosphate (GITP), guanosine triphosphate (GTP), progesterone and nitroimidazole embedded nanoparticle (NNP) effectors, while prostaglandin E2 and F2α were used as effectors in cultured Kupffer cells. The results showed that NNPs decreased adenylate cyclase specific activity in a dose-dependent manner after preincubation of hepatocytes with NNPs. The NNPs stimulated adenylate cyclase activities in hepatocytes were evaluated based on measurement of cyclic adenosine monophosphate (cAMP). The stimulatory effects of NNPs on adenylate cyclase were independent of the presence of GTP and may have been due to a direct effect on the catalytic subunit of adenylate cyclase. In addition, basal cAMP generation in hepatocyte cells was efficiently suppressed by the NNPs. In conclusion, NNPs exerted direct effects on the catalytic subunit of the adenylate cyclase system, and adenylate cyclase was hormone sensitive in liver cells.


Asunto(s)
Adenilil Ciclasas/metabolismo , Hepatocitos/enzimología , Macrófagos del Hígado/enzimología , Nanocápsulas/química , Nitroimidazoles/toxicidad , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/patología , Nanocápsulas/toxicidad , Nanocápsulas/ultraestructura , Fármacos Sensibilizantes a Radiaciones/toxicidad
20.
Adv Mater ; 28(5): 898-904, 2016 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-26635317

RESUMEN

DNA-driven hierarchical core-satellite nanostructures with plasmonic gold nanorod dimers and upconversion nanoparticles are fabricated. Once the core-satellite structure is activated, combined photothermal therapy and photodynamic therapy are carried out under the guidance of upconversion luminesce, T1 -weighted magnetic resonance, photoacoustics, and computed tomography imaging of tumors in vivo, which exhibit the multifunctional biological applications of the DNA-based self-assemblies.


Asunto(s)
Nanotubos/química , Neoplasias/terapia , Fototerapia , Acrilatos/química , Animales , Supervivencia Celular/efectos de los fármacos , Clorofilidas , Exorribonucleasas/química , Oro/química , Células HeLa , Humanos , Rayos Infrarrojos , Ratones , Ratones Desnudos , Nanopartículas/química , Nanotubos/toxicidad , Neoplasias/diagnóstico por imagen , Poliestirenos/química , Porfirinas/química , Porfirinas/uso terapéutico , Porfirinas/toxicidad , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/toxicidad , Tomografía Computarizada por Rayos X , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...