Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
BMJ Open ; 14(1): e072443, 2024 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-38199627

RESUMEN

INTRODUCTION: Peri-implantitis, a common biological complication of dental implant, has attracted considerable attention due to its increasing prevalence and limited treatment efficacy. Previous studies have reported several risk factors associated with the onset of peri-implantitis (eg, history of periodontitis, poor plaque control and smoking). However, inadequate data are available on the association between these risk factors and successful outcome after peri-implantitis therapy. This prospective cohort study aims to identify the local and systemic predictive factors for the treatment success of peri-implantitis. METHODS AND ANALYSIS: A single-centre cohort study will be conducted by recruiting 275 patients diagnosed with peri-implantitis. Sociodemographic variables, healthy lifestyles and systemic disorders will be obtained using questionnaires. In addition, clinical and radiographic examinations will be conducted at baseline and follow-up visits. Treatment success is defined as no bleeding on probing on more than one point, no suppuration, no further marginal bone loss (≥0.5 mm) and probing pocket depth ≤5 mm at the 12-month follow-up interval. After adjustment for age, sex and socioeconomic status, potential prognostic factors related to treatment success will be identified using multivariable logistic regression models. ETHICS AND DISSEMINATION: This cohort study in its current version (2.0, 15 July 2022) is in accordance with the Declaration of Helsinki and was approved by the Ethics Committee of Stomatological Hospital, Southern Medical University (EC-CT-(2022)34). The publication will be on behalf of the study site. TRIAL REGISTRATION NUMBER: ChiCTR2200066262.


Asunto(s)
Enfermedades Óseas Metabólicas , Periimplantitis , Humanos , Estudios de Cohortes , Periimplantitis/diagnóstico por imagen , Periimplantitis/etiología , Periimplantitis/terapia , Estudios Prospectivos , Resultado del Tratamiento
2.
Gynecol Oncol ; 134(1): 190-5, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24811574

RESUMEN

OBJECTIVE: To review recent studies about the application of aromatase inhibitors in endometrial carcinoma and the benefits and challenges of aromatase inhibitors in this regard. METHODS: Relevant studies and manuscripts were searched for in Pubmed using the following terms, either alone or in combination: aromatase, aromatase inhibitors, letrozole, anastrozole, endometrial cancer, breast cancer, endocrine, therapy, and side effects. RESULTS: Endometrial carcinoma is one of the most pervasive gynecological malignancies. Type I endometrial carcinoma is estrogen-dependent. Recent studies have demonstrated that aromatase inhibitors, which interfere with estrogen biosynthesis by inhibiting the activity of aromatase, can be used to treat endometrial carcinoma and its precancerous lesions to some extent. In early-stage endometrial carcinoma or atypical hyperplasia, a precancerous lesion of endometrial carcinoma, the effects of aromatase inhibitors were promising. However, in advanced or recurrent endometrial carcinoma, the application of aromatase inhibitors cannot solve the problem evidently. In addition, these inhibitors have limitations, like side effects and drug resistance. The need for a new generation of inhibitors with higher specificity and fewer side effects should be studied further. CONCLUSIONS: Aromatase inhibitors show promise in the therapy of endometrial carcinoma, especially the early stage. Further studies should be conducted to develop next-generation aromatase inhibitors with higher specificity and fewer side effects.


Asunto(s)
Inhibidores de la Aromatasa/uso terapéutico , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/enzimología , Femenino , Humanos
3.
Gynecol Oncol ; 129(3): 505-12, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23438672

RESUMEN

OBJECTIVE: Visfatin, a newly discovered adipocytokine, is thought to play a role in the pathogenesis of metabolic-syndrome-related cancers. The aim of this study was to assess the clinical significance of serum levels and tissue expression of visfatin in relation to endometrial cancer (EC). METHODS: A total of 234 EC patients were included in this study. Serum visfatin, metabolic and anthropometric parameters were measured in EC patients and controls. Serum visfatin levels were detected using ELISA. Tissue expression of visfatin was analyzed using immunohistochemistry in tissue microarrays. The correlation between clinicopathological variables and visfatin in EC tissues and the prognostic value of visfatin for overall survival was evaluated. RESULTS: Serum levels of visfatin were significantly higher in EC patients than in controls (P<0.05). In univariate and multivariate logistic regression models, a positive association between EC and serum visfatin, BMI, waist-to-hip ratio, diabetes, and hypertension was evident (P<0.05). Visfatin expression was significantly higher in EC tissue than in normal endometrial tissue (P=0.001). Moreover, serum visfatin levels were significantly positively correlated with tissue expression of visfatin in EC patients (P<0.05). High visfatin expression in EC tissues was significantly associated with advanced FIGO stage (P=0.016) and myometrial invasion ≥1/2 (P=0.023). The overall survival rate of EC patients was significantly higher in the group with negative visfatin expression than with positive visfatin expression (P=0.035). CONCLUSIONS: Visfatin is a potential serum biomarker and prognostic factor for EC that may indicate high risk for EC and EC progression. It may also be a novel potential therapeutic target for EC.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Citocinas/biosíntesis , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Nicotinamida Fosforribosiltransferasa/biosíntesis , Biomarcadores de Tumor/sangre , Estudios de Casos y Controles , Citocinas/sangre , Neoplasias Endometriales/sangre , Neoplasias Endometriales/enzimología , Femenino , Humanos , Persona de Mediana Edad , Nicotinamida Fosforribosiltransferasa/sangre , Curva ROC , Factores de Riesgo
4.
Tumour Biol ; 33(5): 1349-62, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22492237

RESUMEN

This study evaluated the effects of a mammalian target of mTOR inhibitor everolimus alone or in combination with trastuzumab on stem cells from HER2-overexpressing primary breast cancer cells and the BT474 breast cancer cell line in vitro and in vivo. For the in vitro studies, we sorted ESA(+)CD44(+)CD24(-/low) cells as stem cells from primary breast cancer cells and BT474 cells using flow cytometry. The MTT assay was used to quantify the inhibitory effect of the drugs on total cells and stem cells specifically. Stem cell apoptosis, cell cycle distributions, and their tumorigenicity after treatment were investigated by flow cytometry or soft agar colony formation assays. For the in vivo studies, BALB/c mice were injected with BT474 stem cells, and the different treatments were administered. After necropsy, the expression of Ki67, CD31, AKT1, and phospho-AKT (Thr308) was analyzed by immunohistochemistry. For the in vitro studies, Treatment with everolimus resulted in stem cell growth inhibition in a dose-dependent manner. The combination of everolimus with trastuzumab was more effective at inhibiting cell growth (P < 0.001) and tumorigenicity (P < 0.001) compared with single-agent therapy. In addition, an increase in G1 cell cycle arrest and an increased population of cells in early apoptosis were seen in the combination treatment group compared with either of the single-agent groups (P < 0.01). For the in vivo studies, everolimus plus trastuzumab therapy was much more effective at reducing tumor volume in mice compared with either single agent alone (P < 0.05). Compared with everolimus alone, the combination of everolimus and trastuzumab reduced the expression of Ki67, AKT1, and phospho-AKT (Thr308) (P < 0.05). We conclude that everolimus has effective inhibitory effects on HER2-overexpressing stem cells in vitro and vivo. Everolimus plus trastuzumab is a rational combination treatment that may be promising in human clinical trials.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Apoptosis , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Everolimus , Femenino , Humanos , Concentración 50 Inhibidora , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-2/antagonistas & inhibidores , Sirolimus/administración & dosificación , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Trastuzumab , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Gynecol Oncol ; 125(3): 751-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22449736

RESUMEN

OBJECTIVE: To review the role played by insulin resistance in the development of endometrial cancer. METHODS: Relevant manuscripts and studies were searched on Medline using the terms endometrial cancer, insulin resistance, obesity, adipokine, C-peptide, leptin, adiponectin, plasminogen activator inhibitor-1, insulin, PI3K/Akt, Ras/MAPK and metformin alone or in combination. RESULTS: Epidemiological studies have shown that insulin resistance is an important potential risk factor of endometrial cancer, and several research studies have been undertaken to determine the mechanism underlying its link to this malignant disease. Risk factors of insulin resistance, such as the inflammatory mediators, adipokines adiponectin, leptin and plasminogen activator inhibitor-1 and excessive androgen are also risk factors of endometrial cancer. High levels of insulin induced by insulin resistance have been found to exert direct and indirect effects that contribute to the development of endometrial cancer. Insulin directly promotes cell proliferation and survival through the PI3K/Akt and Ras/MAPK pathways. Moreover, the network among insulin, estrogen and insulin-like growth factor-1 also contributes to the development of endometrial cancer. Indirectly, insulin leads to changes in sex hormone levels, including increases in the levels of estrogen. Additionally, a small number of studies suggested that metformin, an insulin-sensitizing agent, has therapeutic potential for endometrial cancer. CONCLUSIONS: This evidence suggests that insulin resistance plays a central role in endometrial cancer development. Understanding the relationship between insulin resistance and endometrial cancer may supply new ideas to fight this malignancy. Furthermore, combating insulin resistance may be a useful preventive and therapeutic strategy for endometrial cancer.


Asunto(s)
Neoplasias Endometriales/metabolismo , Resistencia a la Insulina , Animales , Femenino , Humanos , Factores de Riesgo
6.
Eur J Cancer ; 48(10): 1581-92, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22420943

RESUMEN

Recent evidence has suggested that breast cancer contains a rare population of cells called cancer stem cells (CSCs), which have an extensive self-renewal ability and contribute to metastasis and therapeutic resistance. This study evaluated the in vitro and in vivo effects of RAD001 (Everolimus) alone or in combination with docetaxel on stem cells from primary breast cancer cells and two breast cancer cell lines (MCF-7 and MDA-MB-231). In In vitro studies, we sorted ESA(+)CD44(+)CD24(-/low) cells as stem cells using flow cytometry from primary breast cancer cells, MCF-7 and MDA-MB-231 cell lines. MTT assays were used to quantify the inhibitory effect of the drugs on total cells and stem cells. Apoptosis and the cell cycle distributions of stem cells were examined by flow cytometry. The tumourigenicity of stem cells after treatment was investigated by soft agar colony formation assays. In In vivo studies, the BALB/c mice were injected with MDA-MB-231 stem cells and the different treatments were administered. After necropsy, the expression of Ki67, CD31, AKT1, and phospho-AKT (Thr308) was analysed by immunohistochemistry. In In vitro studies, all three populations of stem cells were resistant to the standard treatment doses of docetaxel compared with total cells treated with the same drug. Treatment with RAD001 resulted in growth inhibition of all stem cells in a dose-dependent manner. An additive growth inhibitory effect of the combination treatment on the three stem cells was observed in in vitro compared with treatment with RAD001 alone (P<0.001). In addition, an increase in G2/M cell cycle arrest and an increased population of cells in early apoptosis were seen in the combination treatment group compared with either single-agent group (P<0.01). In vivo, the volumes of the xenograft tumours significantly decreased in RAD001 alone group compared to control group (P=0.008), and RAD001 plus docetaxel therapy was much more effective at reducing tumour volume in mice compared with either single-agent alone (P<0.05). Compared with RAD001 alone, the combination of RAD001 and docetaxel reduced the expression of Ki67, CD31, AKT1 and phospho-AKT (Thr308) (P<0.05). We conclude that the combination treatment of RAD001 and docetaxel can inhibit the growth of stem cells in vitro and in vivo by inhibiting cell proliferation, inducing apoptosis, cell cycle arrest and reducing the expression of Ki67, CD31, AKT1 and phospho-AKT (Thr308). These data indicate that combination treatment with RAD001 and docetaxel may represent an effective therapy for breast cancer. However, further studies are required to evaluate the drug interaction between RAD001 and docetaxel in the clinical setting.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Sirolimus/análogos & derivados , Células Madre/citología , Taxoides/administración & dosificación , Animales , Línea Celular Tumoral , Proliferación Celular , Docetaxel , Relación Dosis-Respuesta a Droga , Everolimus , Femenino , Humanos , Inmunohistoquímica/métodos , Inmunosupresores/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Sirolimus/administración & dosificación , Células Madre/efectos de los fármacos , Sales de Tetrazolio/farmacología , Tiazoles/farmacología
7.
Cancer Lett ; 322(2): 223-31, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22459351

RESUMEN

The involvement of insulin in endometrial carcinoma (EC) was investigated using radioimmunoassay, Western blot, immunoprecipitation, MTT, and Annexin V-FITC/PI assays in tissue samples and cultured cells. Serum levels of insulin, p-p52Shc, p-p46Shc, Shc·Grb2 complexes, p-MEK, p-ERK, and cyclin D1 were elevated in patients with EC. Expression of key proteins in the MEK/ERK pathway, including p-p52Shc, Shc·Grb2 complexes, p-MEK, p-ERK, and cyclin D1, was significantly higher in patients with advanced FIGO stage, high grade, and lymph-node metastasis and correlated positively with serum insulin concentration. Insulin promotes Ishikawa 3-H-12 cell proliferation, survival, and invasion, and these effects induced by insulin were significantly blocked by MEK inhibitor PD98059. Insulin thus promotes EC cell proliferation, survival, and invasion via the MEK/ERK pathway.


Asunto(s)
Neoplasias Endometriales/enzimología , Insulina/metabolismo , Sistema de Señalización de MAP Quinasas , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Insulina/sangre , Insulina/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Invasividad Neoplásica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...