Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cancer ; 18(1): 156, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31694640

RESUMEN

BACKGROUND: Aspartate ß-hydroxylase (ASPH) is silent in normal adult tissues only to re-emerge during oncogenesis where its function is required for generation and maintenance of malignant phenotypes. Exosomes enable prooncogenic secretome delivering and trafficking for long-distance cell-to-cell communication. This study aims to explore molecular mechanisms underlying how ASPH network regulates designated exosomes to program development and progression of breast cancer. METHODS: Stable cell lines overexpressing or knocking-out of ASPH were established using lentivirus transfection or CRISPR-CAS9 systems. Western blot, MTT, immunofluorescence, luciferase reporter, co-immunoprecipitation, 2D/3-D invasion, tube formation, mammosphere formation, immunohistochemistry and newly developed in vitro metastasis were applied. RESULTS: Through physical interactions with Notch receptors, ligands (JAGs) and regulators (ADAM10/17), ASPH activates Notch cascade to provide raw materials (especially MMPs/ADAMs) for synthesis/release of pro-metastatic exosomes. Exosomes orchestrate EMT, 2-D/3-D invasion, stemness, angiogenesis, and premetastatic niche formation. Small molecule inhibitors (SMIs) of ASPH's ß-hydroxylase specifically/efficiently abrogated in vitro metastasis, which mimics basement membrane invasion at primary site, intravasation/extravasation (transendothelial migration), and colonization/outgrowth at distant sites. Multiple organ-metastases in orthotopic and tail vein injection murine models were substantially blocked by a specific SMI. ASPH is silenced in normal adult breast, upregulated from in situ malignancies to highly expressed in invasive/advanced ductal carcinoma. Moderate-high expression of ASPH confers more aggressive molecular subtypes (TNBC or Her2 amplified), early recurrence/progression and devastating outcome (reduced overall/disease-free survival) of breast cancer. Expression profiling of Notch signaling components positively correlates with ASPH expression in breast cancer patients, confirming that ASPH-Notch axis acts functionally in breast tumorigenesis. CONCLUSIONS: ASPH-Notch axis guides particularly selective exosomes to potentiate multifaceted metastasis. ASPH's pro-oncogenic/pro-metastatic properties are essential for breast cancer development/progression, revealing a potential target for therapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas de Unión al Calcio/metabolismo , Exosomas/metabolismo , Proteínas de la Membrana/metabolismo , Oxigenasas de Función Mixta/metabolismo , Proteínas Musculares/metabolismo , Proteoma , Receptores Notch/metabolismo , Animales , Biomarcadores , Comunicación Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Femenino , Genes Reporteros , Xenoinjertos , Humanos , Inmunohistoquímica , Ligandos , Metaloproteinasas de la Matriz/metabolismo , Ratones , Modelos Biológicos , Metástasis de la Neoplasia , Fenotipo , Transducción de Señal
2.
Toxicol Appl Pharmacol ; 359: 55-61, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30244121

RESUMEN

Triple negative breast cancer (TNBC) is associated with poor prognosis and systemic chemotherapy is the only treatment for TNBC. However, development of chemo-resistance remains a major obstacle for TNBC treatment. Paclitaxel-resistance is mainly related to the activation of the Akt signaling pathway and deregulation of apoptotic regulatory proteins. LncRNAs are frequently dysregulated in various malignancies, including breast cancer, facilitating cell proliferation, metastasis and drug resistance. LncRNA H19 is overexpressed in approximately 70% of breast cancer patients, and has been reported to confer chemo-resistance in breast cancer. In the present study, we investigated the expression level of lncRNA H19 in paclitaxel-resistant and paclitaxel-sensitive cell lines. The results showed that the level of lncRNA H19 expression in paclitaxel-resistant cells was significantly higher than that in paclitaxel-sensitive cells, and knockdown of lncRNA H19 might restore chemo-sensitivity in paclitaxel-resistant TNBC by mediating the AKT signaling pathway. Thus, lncRNA H19 might be an efficient therapeutic target in paclitaxel-resistant TNBC treatment.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Proteína Oncogénica v-akt/genética , Paclitaxel/farmacología , ARN Largo no Codificante/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Animales , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Proteína Oncogénica v-akt/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
3.
Hepatology ; 63(4): 1213-26, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26683595

RESUMEN

UNLABELLED: Aspartate ß-hydroxylase (ASPH) is an enzyme overexpressed in human hepatocellular carcinoma (HCC) tumors that participates in the malignant transformation process. We determined if ASPH was a therapeutic target by exerting effects on cellular senescence to retard HCC progression. ASPH knockdown or knockout was achieved by short hairpin RNAs or the CRISPR/Cas9 system, respectively, whereas enzymatic inhibition was rendered by a potent second-generation small molecule inhibitor of ASPH. Alterations of cell proliferation, colony formation, and cellular senescence were evaluated in human HCC cell lines. The potential mechanisms for activating cellular senescence were explored using murine subcutaneous and orthotopic xenograft models. Inhibition of ASPH expression and enzymatic activity significantly reduced cell proliferation and colony formation but induced tumor cell senescence. Following inhibition of ASPH activity, phosphorylation of glycogen synthase kinase 3ß and p16 expression were increased to promote senescence, whereas cyclin D1 and proliferating cell nuclear antigen were decreased to reduce cell proliferation. The mechanisms involved demonstrate that ASPH binds to glycogen synthase kinase 3ß and inhibits its subsequent interactions with protein kinase B and p38 upstream kinases as shown by coimmunoprecipitation. In vivo experiments demonstrated that small molecule inhibitor treatment of HCC bearing mice resulted in significant dose-dependent reduced tumor growth, induced phosphorylation of glycogen synthase kinase 3ß, enhanced p16 expression in tumor cells, and promoted cellular senescence. CONCLUSIONS: We have identified a new mechanism that promotes HCC growth and progression by modulating senescence of tumor cells; these findings suggest that ASPH enzymatic activity is a novel therapeutic target for HCC.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Carcinoma Hepatocelular/enzimología , Senescencia Celular/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Neoplasias Hepáticas/enzimología , Proteínas de la Membrana/metabolismo , Oxigenasas de Función Mixta/metabolismo , Proteínas Musculares/metabolismo , Animales , Carcinoma Hepatocelular/patología , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Glucógeno Sintasa Quinasa 3 beta , Xenoinjertos , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , Fosforilación , Distribución Aleatoria , Sensibilidad y Especificidad , Células Tumorales Cultivadas
4.
Tumour Biol ; 37(1): 943-52, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26260272

RESUMEN

As the most important detoxifying enzymes in liver, glutathione S-transferases (GSTs) can protect hepatocytes against carcinogens. We conducted a large cohort study to investigate the prognostic value of single nucleotide polymorphisms (SNPs) in seven encoding genes of GSTs for hepatocellular carcinoma (HCC). Twelve SNPs were genotyped and correlated with overall survival in 469 HCC patients. The median follow-up time of all patients was 21 (range 3-60) months, and the median survival time was 22 months. By the end of the study, 135 (28.8 %) patients were alive. Only rs4147581 in GSTP1 gene exhibited a significant association with survival of HCC patients (P = 0.006), with its mutant allele bearing a significantly lower risk of death (hazard ratio, 0.71; 95 % confidence interval 0.53-0.90), compared with the homozygous wide-type. A longer median survival time in patients with rs4147581 mutant allele was noticed than those homozygous wide-type (P = 0.03), and there was a marked adverse effect on survival conferred by smoking exposure in these patients. Conclusively, our findings provide supporting evidence for a contributory role of GSTP1 rs4147581 polymorphism in predicting the prognosis of HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidad , Gutatión-S-Transferasa pi/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidad , Polimorfismo de Nucleótido Simple , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Carcinoma Hepatocelular/patología , Femenino , Estudios de Seguimiento , Genotipo , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Factores de Riesgo , Carga Tumoral , Adulto Joven
5.
Cancer Invest ; 34(4): 167-72, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27057601

RESUMEN

PURPOSE: To compare the time-to-treatment failure (TTF), overall survival (OS), overall response rate (ORR), and adverse effects of regimens including nedaplatin- or cisplatin-based chemotherapy for advanced breast cancer (ABC). METHODS: A total of 171 patients with ABC (admission between July 2008 and July 2013) were retrospectively analyzed. Patients received either nedaplatin 75 mg/m(2) (arm N; n = 85) or cisplatin 75 mg/m(2) (arm C; n = 86) in combination with other second-generation chemotherapeutic drugs, such as paclitaxel 175 mg/m(2), docetaxel 75 mg/m(2), gemcitabine 1.25 g/m(2), and navelbine 25 mg/m(2) every 21 days (nedaplatin, cisplatin, paclitaxel, docetaxel on day 1; gemcitabine, navelbine on days 1 and 8). The primary endpoint was TTF in each arm; secondary endpoints were OS, ORR, and toxicity. RESULTS: In the assessable patient population, in arm N, median TTF and OS was 13.87 months (95% CI: 11.55-16.19) and 31.53 months (95% CI: 28.42-34.64), respectively, with an ORR of 48.2%. In arm C, median TTF and OS was 8.7 months (95% CI: 5.82-11.59) and 24.87 months (95% CI: 18.98-30.75), respectively, with an ORR of 37.2%. The occurrence of grades 3 and 4 hematologic toxicity was more frequent (45.9% vs. 25.6%, p = 0.003) in arm N than in arm C. However, grade ≥2 nonhematologic toxicity was less frequent in arm N than in arm C (12.9% vs. 46.5%, p = 2.05 × 10(-7)). CONCLUSIONS: Nedaplatin-based chemotherapy regimen was well tolerated and efficiently improved patients' quality of life characterized by prolonged TTF and OS, with a marginal ORR.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Adulto , Anciano , Cisplatino/administración & dosificación , Cisplatino/efectos adversos , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/efectos adversos
6.
Hepatology ; 60(4): 1302-13, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24954865

RESUMEN

UNLABELLED: Hepatocellular carcinoma (HCC) has a poor prognosis as a result of widespread intra- and extrahepatic metastases. There is an urgent need to understand signaling cascades that promote disease progression. Aspartyl-(asparaginyl)-ß-hydroxylase (ASPH) is a cell-surface enzyme that generates enhanced cell motility, migration, invasion, and metastatic spread in HCC. We hypothesize that inhibition of its enzymatic activity could have antitumor effects. Small molecule inhibitors (SMIs) were developed based on the crystal structure of the ASPH catalytic site followed by computer-assisted drug design. Candidate compounds were tested for inhibition of ß-hydroxylase activity and selected for their capability to modulate cell proliferation, migration, invasion, and colony formation in vitro and to inhibit HCC tumor growth in vivo using orthotopic and subcutaneous murine models. The biological effects of SMIs on the Notch signaling cascade were evaluated. The SMI inhibitor, MO-I-1100, was selected because it reduced ASPH enzymatic activity by 80% and suppressed HCC cell migration, invasion, and anchorage-independent growth. Furthermore, substantial inhibition of HCC tumor growth and progression was observed in both animal models. The mechanism(s) for this antitumor effect was associated with reduced activation of Notch signaling both in vitro and in vivo. CONCLUSIONS: These studies suggest that the enzymatic activity of ASPH is important for hepatic oncogenesis. Reduced ß-hydroxylase activity generated by the SMI MO-I-1100 leads to antitumor effects through inhibiting Notch signaling cascade in HCC. ASPH promotes the generation of an HCC malignant phenotype and represents an attractive molecular target for therapy of this fatal disease.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Proteínas de la Membrana/metabolismo , Oxigenasas de Función Mixta/metabolismo , Animales , Biomarcadores de Tumor/antagonistas & inhibidores , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Furanos/farmacología , Xenoinjertos , Humanos , Técnicas In Vitro , Neoplasias Hepáticas/patología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/efectos de los fármacos , Ratones , Ratones Desnudos , Oxigenasas de Función Mixta/antagonistas & inhibidores , Oxigenasas de Función Mixta/efectos de los fármacos , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/fisiopatología , Receptores Notch/antagonistas & inhibidores , Receptores Notch/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ácidos Sulfónicos/farmacología
7.
J Cell Physiol ; 229(11): 1855-62, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24692224

RESUMEN

Angiotensin II (Ang II), a biologically active peptide of the renin-angiotensin system (RAS), plays an important role in promoting cell migration via Angiotensin II type 1 receptor (AT1R). In this study, we examined the mechanisms by which Ang II affected cell migration in AT1R-positive MDA-MB-231 human breast cancer cells. Ang II increased cell migration and expression of matrix metalloproteinase (MMP)-2,-9 in a dose-dependent manner. Ang II-mediated cell migration was reduced by specific blocking of MMP-2 and MMP-9, as well as with pretreatment with inhibitors of AT1R, phosphatidylinositol 3-kinase (PI3K), Akt, and NF-κB. Similarly, Ang II-mediated expression of MMP-2,-9 was downregulated by pretreatment with inhibitors of AT1R and PI3K. In addition, Ang II treatment significantly induced phosphorylation of PI3K, Akt, and resulted in increased NF-κB activity. These findings suggest that Ang II activates the AT1R/PI3K/Akt pathway, which further activates IKKα/ß and NF-κB, resulting in enhanced expression of MMP-2,-9 and migration in human breast cancer cells. Therefore, targeting Ang II/AT1R/PI3K/Akt/NF-κB signaling could be a novel anti-metastatic therapy for breast cancer.


Asunto(s)
Angiotensina II/farmacología , Neoplasias de la Mama/patología , Movimiento Celular/efectos de los fármacos , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
8.
Tumour Biol ; 35(9): 9371-80, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24946721

RESUMEN

Transforming growth factor-ß1 (TGFß-1) signaling is regulated by endocytotic pathway. To clarify the prognostic value of TGFß-1 and to verify the involvement of endocytosis in drug resistance, we examined the expression of TGFß-1 and Eps15 homology domain 1 (EHD1) in non-small cell lung cancer (NSCLC) and its association with tumor characteristics and survival of patients with NSCLC. Expression of TGFß-1 and EHD1 was evaluated by immunohistochemistry in paraffin sections from 105 NSCLC patients. Overall survival (OS) was analyzed by Kaplan-Meier method, log-rank test, and multivariate Cox proportional hazard regression model. Positive immunostaining of TGFß-1 and EHD1 was detected in 52.38 and 39.05 % of NSCLC samples, respectively. In non-adjuvant chemotherapy-treated group (P = 0.006) and epidermal growth factor receptor (EGFR) (+) group (P = 0.038), patients with TGFß-1 expression had a longer OS. EHD1 negative expression predicted a longer OS (P = 0.003), especially in EGFR (+) (P = 0.006) and adjuvant chemotherapy-treated patients (P = 0.003). NSCLC patients with concurrent positive TGFß-1 and negative EHD1 (combined markers) were significantly correlated with better OS (P = 0.001). American Joint Committee on Cancer (AJCC) status and combined markers were independent prognostic indicators for OS (HR (95 % CI) 1.576 (1.112-2.232), P = 0.011 and HR 0.349 (0.180-0.673), P = 0.002, respectively). We identified concordant TGFß-1 positive and EHD1 negative as a strong favorable prognosis factor in NSCLC. Our results may help us to select and optimize strategies for individualized therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Proteínas de Transporte Vesicular/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/cirugía , Quimioterapia Adyuvante , Terapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/cirugía , Masculino , Persona de Mediana Edad , Análisis Multivariante , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , Pronóstico , Modelos de Riesgos Proporcionales
9.
Tumour Biol ; 35(12): 12707-12, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25217323

RESUMEN

Excision repair cross-complementing gene 1 (ERCC1) protein is proposed as a predictor for cisplatin efficacy in patients with non-small cell lung cancer (NSCLC). However, recent studies declare that ERCC1 is not associated with the response of platinum-based chemotherapy or clinical outcomes. The purpose of this study is to assess whether ERCC1 expression level is linked to cisplatin sensitivity and clinical outcomes in resected NSCLC patients. Paraffin-embedded cancer samples from 112 patients were used for immunohistochemical staining. Cancer cells isolated from fresh tumor tissues were used to determine the sensitivity to cisplatin by MTT assay. The association between ERCC1 expression and cisplatin sensitivity was tested by Spearman's rho test. The correlation of ERCC1 expression with clinicopathologic parameters was evaluated by the chi-square tests. The relationship between variables and survival was assessed by log-rank test. Overall survival (OS) and disease-free survival (DFS) curves were plotted by the Kaplan-Meier method. Cox proportional hazards model was used for multivariate analysis of survival. ERCC1 expression was significantly correlated with the sensitivity of cisplatin in vitro (p < 0.01, r = 0.37). ERCC1 was not associated with OS (p = 0.17) or DFS (p = 0.13) in patients with resected NSCLC. ERCC1 is not a sensible marker for the choice of treatment in clinical patients with resected NSCLC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Adulto , Anciano , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Cisplatino/administración & dosificación , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos/genética , Endonucleasas/metabolismo , Femenino , Expresión Génica , Humanos , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Factores de Riesgo , Resultado del Tratamiento
10.
Tumour Biol ; 35(11): 10841-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25081377

RESUMEN

Cryoablation and surgery achieve similar removal rates for some colorectal cancer (CRC) liver metastasis removal, and systemic chemotherapy is accepted as the most important approach to improving overall survival (OS) in CRC patients with liver metastases. We aimed to evaluate the potential benefit of cryoablation plus chemotherapy in CRC patients with liver metastases. We retrospectively analyze 63 patients of CRC liver metastasis. There were 32 patients in group A, who have received cryoablation plus chemotherapy, and there were 31 patients in group B, who have received chemotherapy alone. We mainly observe the 2-year survival, the quality of life (QOL), and adverse effects. Patients in group A had a higher 2-year survival rate, better OS, better QOL, and better treatment response than patients in group B. Two-year survival rates were 71.9 and 51.6%,respectively, in group A and group B. The negative conversion rates of carcinoembryonic antigen and carbohydrate antigen 19-9 (CA199) were 57.1 and 61.5%, respectively, in group A, and 22.2 and 30%, respectively, in group B. The tumor shrinkage (a tumor volume reduction of ≥ 30%) rates were 62.5 and 22.6%, respectively, in groups A and B. Performance status remained stable or improved in 16 patients (50%) in group A and eight patients (25.81%) in group B. Cryoablation in combination with chemotherapy may increase the 2-year survival rate and improve QOL in CRC patients with liver metastasis.


Asunto(s)
Adenocarcinoma Mucinoso/terapia , Adenocarcinoma/terapia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Colorrectales/terapia , Criocirugía , Neoplasias Hepáticas/terapia , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Adenocarcinoma Mucinoso/mortalidad , Adenocarcinoma Mucinoso/secundario , Adulto , Anciano , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Terapia Combinada , Femenino , Estudios de Seguimiento , Hepatectomía , Humanos , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Calidad de Vida , Estudios Retrospectivos , Tasa de Supervivencia
11.
Helicobacter ; 19(1): 9-16, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24118166

RESUMEN

BACKGROUND: Helicobacter pylori (H. pylori) infection is a major risk factor for gastric cancer (GC); however, only a minority of infected individuals develops GC. We aim to assess the association between serostatus of antibody against H. pylori flagellin A (FlaA) and risk of GC and to evaluate the value of serum FlaA antibody as a novel screening biomarker for GC risk. METHODS: A hospital-based case-control study including 232 cases and 264 controls was conducted. Logistic regression was adopted to analyze the association between the serostatus of FlaA antibody and risk of GC. Serum FlaA antibody was measured by an enzyme-linked immunosorbent assay (ELISA). Receiver operating characteristic (ROC) curve was used to evaluate the screening efficacy and to identify a cutoff point of serum FlaA antibody level. RESULTS: Helicobacter pylori infection was associated with an increased risk of GC (p = .007). A positive association between serum FlaA antibody and GC risk was observed in overall subjects and H. pylori-positive subjects (OR [95% CI]: 6.8 [4.3-10.7] and 6.9 [3.6-13.4], respectively; p < .001). The seropositivity of FlaA antibody was strongly related to GC risk in a dose-dependent manner (p for trend < .001). The optimal cutoff value (OD) was 0.1403, providing a sensitivity of 74.1% and a specificity of 64.4%. The area under the ROC curve (AUC) was 0.74 in overall subjects and 0.73 in H. pylori-positive subjects, respectively. CONCLUSIONS: FlaA was an independent risk factor for H. pylori-related GC. Serum FlaA antibody may serve as a novel noninvasive biomarker for early detection of GC.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Biomarcadores de Tumor/sangre , Flagelina/inmunología , Infecciones por Helicobacter/complicaciones , Helicobacter pylori/inmunología , Neoplasias Gástricas/diagnóstico , Anciano , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Curva ROC , Medición de Riesgo , Sensibilidad y Especificidad
12.
Front Nutr ; 11: 1411206, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38873569

RESUMEN

Objectives: This study aimed to describe the trends of urine lead among US adults aged ≥45 years and to explore its association with all-cause and disease-specific mortality. Methods: This study enrolled 9,669 participants from the National Health and Nutrition Examination Survey, 1999-2018. Trends in urine lead were described by logistic regression analysis using the survey cycle as a continuous variable. Cox proportional hazard regression analyses were used to quantify the association between urine lead and mortality. Results: There was an obvious decline in urine lead concentrations from 1.203 µg/L (95% confidence interval [CI]: 1.083-1.322) in 1999-2000 to 0.478 µg/L (95% CI: 0.433-0.523) in 2017-2018, and this decline was statistically significant (P < 0.001). Referring to the first tertile of urine lead concentrations, risk magnitude for all-cause mortality was significantly and linearly increased after adjustment (P = 0.026 and 0.020 for partially and fully adjusted models, respectively), and significance was attained for the comparison of the third vs. first tertile after full adjustment (hazard ratio [HR]: 1.17, 95% CI: 1.01 to 1.35). Treating urine lead continuously, the risk for all-cause mortality was statistically significant (HR: 1.18 and 1.19, 95% CI: 1.01 to 1.39 and 1.00 to 1.40 for partially and fully adjusted models). For cardiovascular disease-specific and cancer-specific mortality, there was no hint of statistical significance. Conclusions: Our findings indicated that urine lead exhibited a declining trend from 1999-2000 to 2017-2018 in US adults aged ≥45 years, and high urine lead was a significant and independent risk factor for all-cause mortality.

13.
JAMA Netw Open ; 7(6): e2415051, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38837158

RESUMEN

Importance: Obesity, especially visceral obesity, is an established risk factor associated with all-cause mortality. However, the inadequacy of conventional anthropometric measures in assessing fat distribution necessitates a more comprehensive indicator, body roundness index (BRI), to decipher its population-based characteristics and potential association with mortality risk. Objective: To evaluate the temporal trends of BRI among US noninstitutionalized civilian residents and explore its association with all-cause mortality. Design, Setting, and Participants: For this cohort study, information on a nationally representative cohort of 32 995 US adults (age ≥20 years) was extracted from the National Health and Nutrition Examination Survey (NHANES) from 1999 to 2018 and NHANES Linked Mortality File, with mortality ascertained through December 31, 2019. Data were analyzed between April 1 and September 30, 2023. Exposures: Biennial weighted percentage changes in BRI were calculated. Restricted cubic spline curve was used to determine optimal cutoff points for BRI. Main Outcome and Measures: The survival outcome was all-cause mortality. Mortality data were obtained from the Centers for Disease Control and Prevention website and linked to the NHANES database using the unique subject identifier. Weibull regression model was adopted to quantify the association between BRI and all-cause mortality. Results: Among 32 995 US adults, the mean (SD) age was 46.74 (16.92) years, and 16 529 (50.10%) were women. Mean BRI increased gradually from 4.80 (95% CI, 4.62-4.97) to 5.62 (95% CI, 5.37-5.86) from 1999 through 2018, with a biennial change of 0.95% (95% CI, 0.80%-1.09%; P < .001), and this increasing trend was more obvious among women, elderly individuals, and individuals who identified as Mexican American. After a median (IQR) follow-up of 9.98 (5.33-14.33) years, 3452 deaths (10.46% of participants) from all causes occurred. There was a U-shaped association between BRI and all-cause mortality, with the risk increased by 25% (hazard ratio, 1.25; 95% CI, 1.05-1.47) for adults with BRI less than 3.4 and by 49% (hazard ratio, 1.49; 95% CI, 1.31-1.70) for those with BRI of 6.9 or greater compared with the middle quintile of BRI of 4.5 to 5.5 after full adjustment. Conclusions and Relevance: This national cohort study found an increasing trend of BRI during nearly 20-year period among US adults, and importantly, a U-shaped association between BRI and all-cause mortality. These findings provide evidence for proposing BRI as a noninvasive screening tool for mortality risk estimation, an innovative concept that could be incorporated into public health practice pending consistent validation in other independent cohorts.


Asunto(s)
Encuestas Nutricionales , Humanos , Femenino , Masculino , Adulto , Estados Unidos/epidemiología , Persona de Mediana Edad , Mortalidad/tendencias , Estudios de Cohortes , Anciano , Causas de Muerte/tendencias , Factores de Riesgo , Índice de Masa Corporal , Obesidad/mortalidad , Obesidad/epidemiología , Adulto Joven
14.
Carcinogenesis ; 33(7): 1384-90, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22523087

RESUMEN

Four loci have been associated with pancreatic cancer through genome-wide association studies (GWAS). Pathway-based analysis of GWAS data is a complementary approach to identify groups of genes or biological pathways enriched with disease-associated single-nucleotide polymorphisms (SNPs) whose individual effect sizes may be too small to be detected by standard single-locus methods. We used the adaptive rank truncated product method in a pathway-based analysis of GWAS data from 3851 pancreatic cancer cases and 3934 control participants pooled from 12 cohort studies and 8 case-control studies (PanScan). We compiled 23 biological pathways hypothesized to be relevant to pancreatic cancer and observed a nominal association between pancreatic cancer and five pathways (P < 0.05), i.e. pancreatic development, Helicobacter pylori lacto/neolacto, hedgehog, Th1/Th2 immune response and apoptosis (P = 2.0 × 10(-6), 1.6 × 10(-5), 0.0019, 0.019 and 0.023, respectively). After excluding previously identified genes from the original GWAS in three pathways (NR5A2, ABO and SHH), the pancreatic development pathway remained significant (P = 8.3 × 10(-5)), whereas the others did not. The most significant genes (P < 0.01) in the five pathways were NR5A2, HNF1A, HNF4G and PDX1 for pancreatic development; ABO for H.pylori lacto/neolacto; SHH for hedgehog; TGFBR2 and CCL18 for Th1/Th2 immune response and MAPK8 and BCL2L11 for apoptosis. Our results provide a link between inherited variation in genes important for pancreatic development and cancer and show that pathway-based approaches to analysis of GWAS data can yield important insights into the collective role of genetic risk variants in cancer.


Asunto(s)
Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Neoplasias Pancreáticas/genética , Estudios de Casos y Controles , Humanos , Polimorfismo de Nucleótido Simple
15.
Mol Carcinog ; 51(6): 491-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21681824

RESUMEN

DNA repair plays a critical role in human cancers. We hypothesized that DNA mismatch repair gene variants are associated with risk of pancreatic cancer. We retrospectively genotyped 102 single-nucleotide polymorphisms (SNPs) of 13 mismatch repair related genes in 706 patients with pancreatic cancer and 706 cancer-free controls using the mass spectroscopy-based MassArray method. Association of genotype with pancreatic cancer risk was tested by multivariate logistic regression models. A significance level of P ≤ 0.0015 was set at the false discovery rate (FDR) <1% using the Beta-Uniform Mixture method. We found 28 SNPs related to altered pancreatic cancer risk (P < 0.05). Adjusting for multiple comparisons, MGMT I143V AG/GG, PMS2 IVS1-1121C > T TC/TT, and PMS2L3 Ex1 + 118C > T CT/TT genotypes showed significant main effects on pancreatic cancer risk at FDR <1% with OR (95% CI) of 0.60 (0.46-0.80), 1.44 (1.14-1.81), and 5.54 (2.10-14.61), respectively (P ≤ 0.0015). To demonstrate genotype-phenotype association, we measured O(6)-ethylguanosine (O(6)-EtGua) adduct levels in vitro by immunoslot blot assay in lymphocytes treated with N-ethyl-N-nitrosourea (ENU) in 297 case/control subjects. MGMT I143V GG, MGMT K178R GG, MSH6 G39E AG/AA, PMS2L3 IVS3 + 9A > G GA and TP73 IVS1-7449G > C CG/CC genotypes correlated with a higher level of ENU-induced DNA adducts. Haplotypes of MGMT, MSH6, PMS2, PMS2L3, and TP73 were significantly associated with pancreatic cancer risk (P ≤ 0.0015). Our findings suggest that mismatch repair gene variants may affect susceptibility to pancreatic cancer.


Asunto(s)
Reparación de la Incompatibilidad de ADN/genética , Predisposición Genética a la Enfermedad , Neoplasias Pancreáticas/genética , Polimorfismo de Nucleótido Simple , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Genotipo , Humanos , Masculino , Persona de Mediana Edad , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Factores de Riesgo
16.
Mol Biol Rep ; 39(3): 2615-24, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21667251

RESUMEN

The association between the polymorphic CAG repeat in androgen receptor gene (AR) and prostate cancer susceptibility has been studied extensively. However, the results are contradictory. The purpose of our meta-analysis was to investigate whether CAG repeat related to prostate cancer risk and had genetic heterogeneity across different geographic regions and study designs. Random-effects model was performed irrespective of between-study heterogeneity. Data and study quality were assessed in duplicate. Publication bias was assessed by the fail-safe number and Egger's test. There were 16 (patients/controls: 2972/3792), 19 (3835/4908) and 12 (3372/2631) study groups for comparisons of ≥ 20, 22 and 23 repeats of CAG sequence, respectively. Compared with CAG repeat <20, 22 or 23, carriers of ≥ 20, 22 or 23 repeats had 21% (95% CI: 0.61-1.02; P = 0.076), 5% (95% CI: 0.81-1.11; P = 0.508) and 5% (95% CI: 0.76-1.20; P = 0.681) decreased risk of prostate cancer. After classifying studies by geographic areas, carriers of ≥ 20 repeats had 11% decreased risk in populations from USA, 53% from Europe, and 20% from Asia (P > 0.05), whereas comparison of ≥ 23 repeats with others generated a significant prediction in European populations (OR = 1.17; P = 0.039). Stratification by study designs revealed no material changes in risk estimation. Meta-regression analysis found no significant sources of between-study heterogeneity for age, study design and geographic region for all comparisons. There was no identified publication bias. Taken together, our results demonstrated that AR CAG repeat polymorphism with ≥ 20 repeats might confer a protective effect among the prostate cancer patients with 45 years older but not all the prostate cancer patients.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Polimorfismo Genético/genética , Neoplasias de la Próstata/epidemiología , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Repeticiones de Trinucleótidos/genética , Factores de Edad , Demografía , Humanos , Masculino , Modelos Estadísticos , Sesgo de Publicación , Análisis de Regresión , Proyectos de Investigación
17.
Am J Cancer Res ; 12(9): 4343-4360, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36225636

RESUMEN

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death attributed to high frequency of metastasis and multiple drug resistance. We aim to examine the underlying molecular mechanism and to seek potential strategies to reverse primary/acquired resistance to regorafenib. Topoisomerase IIα (TOP2A) is critical for tumorigenesis and carcinogenesis. Clinically, high-TOP2A expression was correlated to shorter overall survival (OS) of patients, but its role in drug resistance of HCC remains unknown. Here, we screened the expression profiling of TOP2A in HCC and identified TOP2A as an upregulated gene involved in the resistance to regorafenib. Sustained exposure of HCC cells to regorafenib could upregulate the expression of TOP2A. Silencing TOP2A enhanced HCC cells' sensitivity to regorafenib. TOP2A inhibition by doxorubicin or epirubicin synergized with regorafenib to suppress the growth of sorafenib-resistant HCC tumors that possessed the sorafenib-resistant features both in vitro and in vivo. Thus, targeting TOP2A may be a promising therapeutic strategy to alleviate resistance to regorafenib and thus improving the efficacy of HCC treatment.

18.
J Exp Clin Cancer Res ; 41(1): 132, 2022 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-35392977

RESUMEN

BACKGROUND: Interactions between tumor and microenvironment determine individual response to immunotherapy. Triple negative breast cancer (TNBC) and hepatocellular carcinoma (HCC) have exhibited suboptimal responses to immune checkpoint inhibitors (ICIs). Aspartate ß-hydroxylase (ASPH), an oncofetal protein and tumor associated antigen (TAA), is a potential target for immunotherapy. METHODS: Subcutaneous HCC and orthotopic TNBC murine models were established in immunocompetent BALB/c mice with injection of BNL-T3 and 4 T1 cells, respectively. Immunohistochemistry, immunofluorescence, H&E, flow cytometry, ELISA and in vitro cytotoxicity assays were performed. RESULTS: The ASPH-MYC signaling cascade upregulates PD-L1 expression on breast and liver tumor cells. A bio-nanoparticle based λ phage vaccine targeting ASPH was administrated to mice harboring syngeneic HCC or TNBC tumors, either alone or in combination with PD-1 blockade. In control, autocrine chemokine ligand 13 (CXCL13)-C-X-C chemokine receptor type 5 (CXCR5) axis promoted tumor development and progression in HCC and TNBC. Interactions between PD-L1+ cancer cells and PD-1+ T cells resulted in T cell exhaustion and apoptosis, causing immune evasion of cancer cells. In contrast, combination therapy (Vaccine+PD-1 inhibitor) significantly suppressed primary hepatic or mammary tumor growth (with distant pulmonary metastases in TNBC). Adaptive immune responses were attributed to expansion of activated CD4+ T helper type 1 (Th1)/CD8+ cytotoxic T cells (CTLs) that displayed enhanced effector functions, and maturation of plasma cells that secreted high titers of ASPH-specific antibody. Combination therapy significantly reduced tumor infiltration of immunosuppressive CD4+/CD25+/FOXP3+ Tregs. When the PD-1/PD-L1 signal was inhibited, CXCL13 produced by ASPH+ cancer cells recruited CXCR5+/CD8+ T lymphocytes to tertiary lymphoid structures (TLSs), comprising effector and memory CTLs, T follicular helper cells, B cell germinal center, and follicular dendritic cells. TLSs facilitate activation and maturation of DCs and actively recruit immune subsets to tumor microenvironment. These CTLs secreted CXCL13 to recruit more CXCR5+ immune cells and to lyse CXCR5+ cancer cells. Upon combination treatment, formation of TLSs predicts sensitivity to ICI blockade. Combination therapy substantially prolonged overall survival of mice with HCC or TNBC. CONCLUSIONS: Synergistic antitumor efficacy attributable to a λ phage vaccine specifically targeting ASPH, an ideal TAA, combined with ICIs, inhibits tumor growth and progression of TNBC and HCC.


Asunto(s)
Vacunas contra el Cáncer , Carcinoma Hepatocelular , Inmunoterapia , Neoplasias Hepáticas , Neoplasias de la Mama Triple Negativas , Animales , Antígeno B7-H1 , Vacunas contra el Cáncer/inmunología , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/terapia , Humanos , Inhibidores de Puntos de Control Inmunológico , Inmunidad , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/terapia , Ratones , Nanopartículas , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/terapia , Microambiente Tumoral
19.
Oncologist ; 16(1): 61-70, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21212431

RESUMEN

PURPOSE: DNA mismatch repair (MMR) maintains genomic stability and mediates cellular response to DNA damage. We aim to demonstrate whether MMR genetic variants affect overall survival (OS) in pancreatic cancer. MATERIALS AND METHODS: Using the Sequenom method in genomic DNA, we retrospectively genotyped 102 single-nucleotide polymorphisms (SNPs) of 13 MMR genes from 706 patients with pancreatic adenocarcinoma seen at The University of Texas MD Anderson Cancer Center. Association between genotype and OS was evaluated using multivariable Cox proportional hazard regression models. RESULTS: At a false discovery rate of 1% (p ≤ .0015), 15 SNPs of EXO1, MLH1, MSH2, MSH3, MSH6, PMS2, PMS2L3, TP73, and TREX1 in patients with localized disease (n = 333) and 6 SNPs of MSH3, MSH6, and TP73 in patients with locally advanced or metastatic disease (n = 373) were significantly associated with OS. In multivariable Cox proportional hazard regression models, SNPs of EXO1, MSH2, MSH3, PMS2L3, and TP73 in patients with localized disease, MSH2, MSH3, MSH6, and TP73 in patients with locally advanced or metastatic disease, and EXO1, MGMT, MSH2, MSH3, MSH6, PMS2L3, and TP73 in all patients remained significant predictors for OS (p ≤ .0015) after adjusting for all clinical predictors and all SNPs with p ≤ .0015 in single-locus analysis. Sixteen haplotypes of EXO1, MLH1, MSH2, MSH3, MSH6, PMS2, PMS2L3, RECQL, TP73, and TREX1 significantly correlated with OS in all patients (p ≤ .001). CONCLUSION: MMR gene variants may have potential value as prognostic markers for OS in pancreatic cancer patients.


Asunto(s)
Adenocarcinoma/genética , Reparación de la Incompatibilidad de ADN/genética , Neoplasias Pancreáticas/genética , Adenocarcinoma/mortalidad , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/mortalidad , Polimorfismo de Nucleótido Simple , Modelos de Riesgos Proporcionales , Tasa de Supervivencia
20.
Gastroenterology ; 139(2): 464-73, 473.e1-3, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20416304

RESUMEN

BACKGROUND & AIMS: Insulin-like growth factor (IGF)-axis mediated signaling pathways play an important role in pancreatic cancer development and progression. We examined whether IGF-axis gene variants are associated with clinical outcomes in pancreatic cancer. METHODS: We retrospectively genotyped 41 single-nucleotide polymorphisms from 10 IGF-axis genes in 333 patients with localized pancreatic adenocarcinoma and validated the findings in 373 patients with advanced disease. Associations between genotype and overall survival (OS) were evaluated using multivariable Cox proportional hazard regression models. RESULTS: IGF1 *8470T>C, IGF1R IVS2+46329T>C, IGFBP3 A32G, IRS1 G972R in patients with localized disease; IGF1R IVS20-3431A>G, IGF1R T766T, IGFBP3-202A>C, IRS1 IVS1+4315C>G, IRS1 G972R in patients with advanced disease; and IGF1R T766T, IGF2R L252V, IGFBP3 -202A>C, IRS1 IVS1+4315C>G, IRS1 G972R, IRS2 IVS1+5687T>C in all patients were significantly associated with OS (PA, and an IRS2 haplotype predicted worse OS (P

Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/terapia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Polimorfismo de Nucleótido Simple , Transducción de Señal/genética , Somatomedinas/genética , Anciano , Estudios de Casos y Controles , Ensayos Clínicos Fase II como Asunto , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Estimación de Kaplan-Meier , Modelos Logísticos , Masculino , Persona de Mediana Edad , Fenotipo , Modelos de Riesgos Proporcionales , Receptores de Somatomedina/genética , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA