Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Neurol Surg B Skull Base ; 84(6): 609-615, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37854532

RESUMEN

Objectives Primary frontal sinus malignancies (FSMs) are the rarest sinonasal cancers. This study aimed to determine clinicopathologic characteristics of primary FSMs and provide long-term survival outcomes. Design This study is a retrospective review. Setting The study was conducted at a tertiary medical center. Participants Patients who participated in this study were diagnosed with primary FSMs. Main Outcome Measures Median survival time is the primary outcome measure of this study. Results In this series, the median age was 48 years (30-53 years) and all patients were male. There were five cases with squamous cell carcinoma and one with osteosarcoma. All cases presented with locally advanced disease without regional lymphatic metastasis, including five cases of stage III and one case of stage II. The two most common pathways of tumor invasion were as follows: local tumor broke posteriorly through bone wall and invaded dura mater, followed by frontal lobe; local tumor infiltrated downward through the floor of frontal sinus into ethmoid sinus, thereafter invaded laterally orbit and orbital contents. All patients received surgery followed by postoperative radiotherapy at the total doses of 50 to 75.95 Gy. Among them, only one patient underwent R0 resection, the rest of patients underwent R1/R2 resection. With a median survival time of 56 months (32-76 months), two patients receiving R1/R2 resection developed treatment failure and died within 5 years, including one case with local recurrence and one with local recurrence, thereafter distant metastasis. Conclusion The majority of FSMs presented with peripherally invasive progression lesions which led to a high ratio of R1/R2 resection. Surgery combined with postoperative radiotherapy might result in satisfactory efficacy.

2.
Acta Biomater ; 150: 254-264, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35917911

RESUMEN

Since 1995, photodynamic therapy (PDT) has been utilized as an effective method for cancer treatment. However, the residues of photosensitizers in the normal tissues after PDT can be activated by sunlight to cause severe skin phototoxicity, for which currently there are no clinical solutions. As a result, post-PDT patients need to remain out of sunlight for up to five weeks, which produces great living and mental burdens for patients. Herein, we report that a biocompatible porous organic polymer (POP) with average 3.1 nm porosity is able to suppress the skin phototoxicity of clinically used porphyrin-based photodynamic agents (PDAs), including Photofrin, Talaporfin and Hiporfin, through an adsorption-elimination mechanism. Fluorescence titration and dialysis experiments show that POP can adsorb and retain the PDAs at a micromolar concentration. In vivo experiments demonstrate that POP can significantly suppress the skin phototoxicity caused by all the three PDAs without reducing their PDT efficacy. STATEMENT OF SIGNIFICANCE: Up to now, no efficient clinical treatment for the inhibition of post-PDT phototoxicity of clinically used porphyrin-based PDAs is available. In the manuscript, a water-soluble cationic porous organic polymer has been revealed to include three clinically used PDAs. In vivo experiments show that this inclusion remarkably reduces the content of PDAs in mouse skins, leading to significant alleviation of their post-PDT phototoxicity without no negative effect on their PDT efficacy. Thus, this work provides a strategy for overcoming the drawback of clinically used photodynamic agents.


Asunto(s)
Fotoquimioterapia , Porfirinas , Animales , Línea Celular Tumoral , Ratones , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/química , Polímeros , Porosidad , Porfirinas/farmacología
3.
J Mater Chem B ; 10(22): 4163-4171, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35551323

RESUMEN

Phase I-III clinical studies show that aldoxorubicin (AlDox), a prodrug of doxorubicin (Dox), displays reduced cardiotoxicity compared to Dox, but does not demonstrate an overall survival benefit in patients. Here we report that three-dimensional supramolecular organic frameworks (SOFs) can conjugate AlDox through quantitative thiol-maleimide addition to afford two polymeric prodrugs of Dox. The previously established ability of SOFs in overcoming the multidrug resistance of tumor cells is utilized to achieve efficient intracellular delivery of the conjugated AlDox, which releases Dox as an active agent through acid-responsive hydrolysis of the hydrazone bond of AlDox within tumor cells. In vitro and in vivo experiments show that conjugation to SOF significantly improves the antitumor efficacy of AlDox as compared with free AlDox of the identical dose. Moreover, the SOF prodrugs do not show cardiotoxicity, the major superiority of AlDox over Dox. Since free AlDox is conjugated to endogenous albumin in the blood through thiol-maleimide addition to achieve enhanced intracellular delivery and Dox release through acid-responsive hydrazone hydrolysis, SOF conjugation provides a surrogate strategy for prodrug design to gain improved efficacy.


Asunto(s)
Profármacos , Cardiotoxicidad , Doxorrubicina/análogos & derivados , Doxorrubicina/química , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos , Humanos , Hidrazonas , Maleimidas , Polímeros/química , Profármacos/química , Profármacos/farmacología , Compuestos de Sulfhidrilo
4.
Biomaterials ; 284: 121467, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35338941

RESUMEN

Despite that photodynamic therapy (PDT) has been applied for the treatment of cancer and skin diseases for more than two decades, all clinically used photodynamic agents (PDAs) suffer the drawback of skin phototoxicity of PDAs, which requires patients to avoid exposure to natural light for weeks after treatment, but has so far lacked effective suppression methods. Here, we report that three-dimensional diamondoid supramolecular organic frameworks (SOFs), that possess well-defined 2.1-nm porosity, can be used to suppress the skin phototoxicity of Photofrin, HiPorfin and Talaporfin, three porphyrin-based PDAs which clinically receive the most wide applications by injecting SOF after PDT, via an adsorption and retention mechanism. Fluorescence and dynamic light scattering experiments confirm that the SOFs have strong interaction with PDAs, and can adsorb PDAs at a micromolar concentration, whereas dialysis experiments support that the adsorption leads to an important retention effect. In vitro and in vivo experiments reveal that SOFs have high biocompatibility. Studies with healthy and tumor-bearing mouse models demonstrate that, when the PDAs are administrated at a dose comparable with the clinical one, SOF can remarkably suppress sunlight-induced skin phototoxicity, whereas the PDT efficacy of mice treated with SOF post-PDT is maintained. This work provides an efficient strategy for the improvement of the safety of clinically used PDAs.


Asunto(s)
Neoplasias , Fotoquimioterapia , Porfirinas , Animales , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Porfirinas/uso terapéutico , Diálisis Renal
5.
J Mater Chem B ; 10(6): 899-908, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35043828

RESUMEN

Water-soluble three-dimensional supramolecular-organic frameworks (SOFs) and temoporfin (mTHPC) are discovered to form uniform self-assembled nanoparticles. These nanoparticles demonstrate an improved 1O2 generation efficiency due to the reduced aggregation-caused quenching effect. SOFs and self-assembled nanoparticles are biocompatible. Self-assembled nanoparticles display an improved photo cytotoxicity toward four types of human cancer cells. The tumor model in mice shows that self-assembled nanoparticles could efficiently suppress tumor growth in vivo.


Asunto(s)
Nanopartículas , Neoplasias , Fotoquimioterapia , Animales , Mesoporfirinas/uso terapéutico , Ratones , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos
6.
Drug Deliv ; 29(1): 128-137, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34967270

RESUMEN

Chemotherapy is one of the main ways to treat breast cancer clinically. However, the multidrug resistance to anti-tumor drugs limits their clinical use. To overcome these drawbacks, development of drug delivery systems (DDSs) has attracted more and more attention in cancer therapy. At present, the preparation and purification process are complicated for many reported DDSs, while clinic calls for new DDSs that are more convenient for preparation. Here, a new pH-responsive supramolecular organic framework drug delivery complex loading doxorubicin (DOX) is fabricated. Anti-tumor activity of the system in vitro was investigated by cell cytotoxicity, uptake assay, and cell apoptosis analysis. The anti-tumor activity in vivo was investigated by inspecting nude mice body weight, tumor volume, and weight, also a preliminary mechanism probe was conducted by HE and TUNEL staining. The DOX@SOF displayed high stability, good biocompatibility, and pH regulated drug release. At acid condition, the hydrazone bonds would be broken, which result in the dissociation of SOF, and then the drugs would be released from the system. Furthermore, DOX@SOF enhanced cellular internalization. Both in vitro and in vivo experiments reflected that DOX@SOF could enhance the anti-tumor activity of DOX for the MCF-7/ADR tumor cells and tumors. This study provides a highly efficient strategy to prepare stimulus-responsive supramolecular drug delivery complex for treatment of drug-resistant cancer, the results presented inspiring scientific interests in exploring new drug delivery strategy and reversing multi-drug resistance for clinical chemotherapy.


Asunto(s)
Portadores de Fármacos/química , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Química Farmacéutica , Doxorrubicina/administración & dosificación , Liberación de Fármacos , Resistencia a Antineoplásicos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Distribución Aleatoria , Propiedades de Superficie , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Drug Deliv ; 29(1): 1-9, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34949133

RESUMEN

Chemotherapy is one of the main ways to treat breast cancer clinically. However, the multidrug resistance to anti-tumor drugs limits their clinical use. To overcome these drawbacks, the development of drug delivery systems (DDSs) has attracted more and more attention in cancer therapy. At present, the preparation and purification process are complicated for many reported DDSs, while the clinic calls for new DDSs that are more convenient for preparation. Here a new pH-responsive supramolecular organic framework drug delivery complex loading doxorubicin (DOX) is fabricated. Anti-tumor activity of the system in vitro was investigated by cell cytotoxicity, uptake assay, and cell apoptosis analysis. The anti-tumor activity in vivo was investigated by inspecting nude mice body weight, tumor volume and weight, also a preliminary mechanism probe was conducted by HE and TUNEL staining. The DOX@SOF displayed high stability, good biocompatibility and pH-regulated drug release. At acid condition, the hydrazone bonds would be broken, which result in the dissociation of SOF, and then the drugs would be released from the system. Furthermore, DOX@SOF enhanced cellular internalization. Both in vitro and in vivo experiments reflected that DOX@SOF could enhance the anti-tumor activity of DOX. for the MCF-7/ADR tumor cells and tumors. This study provides a highly efficient strategy to prepare a stimulus-responsive supramolecular drug delivery complex for the treatment of drug-resistant cancer, the results presented inspiring scientific interests in exploring new drug delivery strategies and reversing multi-drug resistance for clinical chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Doxorrubicina/farmacología , Portadores de Fármacos/farmacología , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular , Doxorrubicina/administración & dosificación , Portadores de Fármacos/administración & dosificación , Liberación de Fármacos , Resistencia a Antineoplásicos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ratones Endogámicos BALB C , Ratones Desnudos , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Nutrition ; 78: 110870, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32659681

RESUMEN

OBJECTIVE: The relationship between vitamin intake and the occurrence of cancer is controversial. The aim of this study was to examine the genetic associations between vitamins D, E, and B12 and five cancers (i.e., colorectal cancer, breast cancer, prostate cancer, malignant melanoma, and squamous cell carcinoma). METHODS: This study started from genome-wide association data for three vitamins (N = 11 238) and five cancers (N = 373 316). The study analyzed their associations using Mendelian randomization (MR) methods. Additionally, survival analysis was performed using data from The Cancer Genome Atlas (TCGA) to further evaluate some MR results. RESULTS: MR analysis indicated that intake of vitamins D, E, and B12 is not relevant to the risk for the five cancers (PMR > Bonferroni-corrected P = 0.02). Some of the results were supported by epidemiological observations; some were further supported by survival analysis using TCGA data. CONCLUSION: There is no genetic evidence to support the association between intake of vitamins D, E, and B12 and the risk for colorectal, breast, and prostate cancers, malignant melanoma, and squamous cell carcinoma.


Asunto(s)
Neoplasias , Vitaminas , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Análisis de la Aleatorización Mendeliana , Neoplasias/epidemiología , Neoplasias/genética , Riesgo , Vitamina A
9.
J Am Chem Soc ; 142(7): 3577-3582, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32011130

RESUMEN

Four water-soluble hydrazone-based three-dimensional (3D) flexible organic frameworks FOF-1-4 have been synthesized from a semirigid tetracationic tetraaldehyde and four flexible dihydrazides. 1H NMR spectroscopy indicated the quantitative formation of FOF-1-4 in D2O, while dynamic light scattering experiments revealed that, depending on the concentration, these porous frameworks display hydrodynamic diameters ranging from 50 to 120 nm. The porosity of the frameworks is confirmed by ethanol vapor adsorption experiments of the solid samples as well as the high loading capacity for a 2.3 nm porphyrin guest in water. The new water-soluble frameworks exhibit low cytotoxicity and form inherent pores with diameters of 5.3 or 6.7 nm, allowing rapid inclusion of proteins such as bovine serum albumin and green and orange fluorescent proteins, and efficient delivery of the proteins into normal and cancer cells. Flow cytometric analysis reveals percentages of the delivered cells up to 99.8%.


Asunto(s)
Estructuras Metalorgánicas/química , Proteínas/química , Etanol/química , Hidrazonas/química , Resonancia Magnética Nuclear Biomolecular , Dispersión de Radiación , Solubilidad , Espectrofotometría Infrarroja , Agua/química
10.
J Transl Med ; 17(1): 221, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31296244

RESUMEN

BACKGROUND: The clinical significance of stem cell therapy in the treatment of dilated cardiomyopathy remains unclear. This systemic appraisal and meta-analysis aimed to assess the efficacy and safety of stem cell therapy in patients with dilated cardiomyopathy. After searching the PubMed, Embase, and Cochrane library databases until November 2017, we conducted a meta-analysis to evaluate the efficacy and safety of stem cell therapy in patients with dilated cardiomyopathy. METHODS: The weighted mean difference (WMD), standard mean difference (SMD), relative risk (RR), and 95% confidence interval (CI) were summarized in this meta-analysis. Both fixed effects and random effects models were used to combine the data. Sensitivity analyses were conducted to evaluate the impact of an individual dataset on the pooled results. RESULTS: A total of eight randomized controlled trials, which involved 531 participants, met the inclusion criteria in this systematic appraisal and meta-analysis. Our meta-analysis showed that stem cell therapy improves left ventricular ejection fraction (SMD = 1.09, 95% CI 0.29 to 1.90, I2 = 92%) and reduces left ventricular end-systolic volume (SMD = - 0.36, 95% CI - 0.61 to - 0.10, I2 = 20.5%) and left ventricular end-diastolic chamber size (SMD = - 0.48, 95% CI - 0.89 to - 0.07, I2 = 64.8%) in patients with dilated cardiomyopathy. However, stem cell therapy has no effect on mortality (RR = 0.72, 95% CI 0.50 to 1.02, I2 = 30.2%) and 6-min-walk test (WMD = 51.52, 95% CI - 24.52 to 127.55, I2 = 94.8%). CONCLUSIONS: This meta-analysis suggests that stem cell therapy improves left ventricular ejection fraction and reduces left ventricular end-systolic volume and left ventricular end-diastolic chamber size in patients with dilated cardiomyopathy. However, future well-designed large studies might be necessary to clarify the effect of stem cell therapy in patients with dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/terapia , Trasplante de Células Madre/efectos adversos , Cardiomiopatía Dilatada/mortalidad , Cardiomiopatía Dilatada/fisiopatología , Diástole , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sesgo de Publicación , Riesgo , Volumen Sistólico , Sístole , Resultado del Tratamiento , Prueba de Paso
11.
Acta Pharmacol Sin ; 39(10): 1613-1621, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29795355

RESUMEN

Hepatocyte growth factor (HGF) alleviates acute and chronic inflammation in experimental inflammatory bowel disease, glomerulonephritis, and airway inflammation. However, the anti-inflammatory effects of HGF on myocardial infarction are not defined. The current study assessed the anti-inflammatory effects of HGF in post-ischemic heart failure. The left anterior descending coronary artery was ligated in rats, and adenovirus containing human HGF (Ad-HGF) or control virus (Ad-GFP) was administered intramyocardially. The quantity of proinflammatory cytokines secreted by cardiomyocytes, such as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-1ß, was evaluated. Cardiac function and LV remodeling were assessed using echocardiography and collagen deposition, respectively. Left ventricular fractional shortening (LVFS) and left ventricular ejection fraction (LVEF) four weeks after injection were significantly increased in Ad-HGF-treated animals compared to the Ad-GFP group. HGF gene therapy improved ventricular geometry with a significantly decreased left ventricular end-diastolic diameter (LVEDD) and markedly reduced myocardial collagen deposition. Treatment with Ad-HGF significantly decreased the mRNA levels of TNF-α, IL-6, and IL-1ß in the non-infarcted region four weeks after injection. Changes of the TNF-α, IL-6, and IL-1ß levels in the non-infarcted region positively correlated with the LVEDD 4 weeks after infarction. Treatment of acute myocardial infarction (AMI) with Ad-HGF in the early stage of MI reduced the pro-inflammatory cytokine levels and preserved cardiac function. These findings indicated that Ad-HGF gene therapy alleviated ventricular remodeling after infarction by reducing inflammation.


Asunto(s)
Infarto de la Pared Anterior del Miocardio/terapia , Insuficiencia Cardíaca/terapia , Factor de Crecimiento de Hepatocito/uso terapéutico , Inflamación/terapia , Adenoviridae/genética , Animales , Infarto de la Pared Anterior del Miocardio/metabolismo , Antiinflamatorios/uso terapéutico , Citocinas/metabolismo , Terapia Genética/métodos , Células HEK293 , Insuficiencia Cardíaca/metabolismo , Factor de Crecimiento de Hepatocito/genética , Humanos , Inflamación/metabolismo , Masculino , Miocardio/metabolismo , Miocardio/patología , Ratas Wistar
12.
World J Gastroenterol ; 18(22): 2850-8, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22719196

RESUMEN

AIM: To investigate whether DNA-dependent activator of interferon-regulatory factors (DAI) inhibits hepatitis B virus (HBV) replication and what the mechanism is. METHODS: After the human hepatoma cell line Huh7 was cotransfected with DAI and HBV expressing plasmid, viral protein (HBV surface antigen and HBV e antigen) secretion was detected by enzyme-linked immunosorbent assay, and HBV RNA was analyzed by real-time polymerase chain reaction and Northern blotting, and viral DNA replicative intermediates were examined by Southern blotting. Interferon regulatory factor 3 (IRF3) phosphorylation and nuclear translocation were analyzed via Western blotting and immunofluorescence staining respectively. Nuclear factor-κB (NF-κB) activity induced by DAI was detected by immunofluorescence staining of P65 and dual luciferase reporter assay. Transwell co-culture experiment was performed in order to investigate whether the antiviral effects of DAI were dependent on the secreted cytokines. RESULTS: Viral protein secretion was significantly reduced by 57% (P < 0.05), and the level of total HBV RNA was reduced by 67% (P < 0.05). The viral core particle-associated DNA was also dramatically down-regulated in DAI-expressing Huh7 cells. Analysis of involved signaling pathways revealed that activation of NF-κB signaling was essential for DAI to elicit antiviral response in Huh7 cells. When the NF-κB signaling pathway was blocked by a NF-κB signaling suppressor (IκBα-SR), the anti-HBV activity of DAI was remarkably abrogated. The inhibitory effect of DAI was independent of IRF3 signaling and secreted cytokines. CONCLUSION: This study demonstrates that DAI can inhibit HBV replication and the inhibitory effect is associated with activation of NF-κB but independent of IRF3 and secreted cytokines.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Virus de la Hepatitis B/crecimiento & desarrollo , Hepatocitos/metabolismo , Hepatocitos/virología , Replicación Viral , Northern Blotting , Southern Blotting , Western Blotting , Línea Celular Tumoral , Técnicas de Cocultivo , Citocinas/metabolismo , ADN Viral/biosíntesis , Proteínas de Unión al ADN/genética , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Células HEK293 , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/inmunología , Hepatocitos/inmunología , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Inhibidor NF-kappaB alfa , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Viral/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA