Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Intervalo de año de publicación
1.
Theranostics ; 14(6): 2290-2303, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38646651

RESUMEN

Background: Neoantigen nanovaccine has been recognized as a promising treatment modality for personalized cancer immunotherapy. However, most current nanovaccines are carrier-dependent and the manufacturing process is complicated, resulting in potential safety concerns and suboptimal codelivery of neoantigens and adjuvants to antigen-presenting cells (APCs). Methods: Here we report a facile and general methodology for nanoassembly of peptide and oligonucleotide by programming neoantigen peptide with a short cationic module at N-terminus to prepare nanovaccine. The programmed peptide can co-assemble with CpG oligonucleotide (TLR9 agonist) into monodispersed nanostructures without the introduction of artificial carrier. Results: We demonstrate that the engineered nanovaccine promoted the codelivery of neoantigen peptides and adjuvants to lymph node-residing APCs and instigated potent neoantigen-specific T-cell responses, eliciting neoantigen-specific antitumor immune responses with negligible systemic toxicity. Furthermore, the antitumor T-cell immunity is profoundly potentiated when combined with anti-PD-1 therapy, leading to significant inhibition or even complete regression of established melanoma and MC-38 colon tumors. Conclusions: Collectively, this work demonstrates the feasibility and effectiveness of personalized cancer nanovaccine preparation with high immunogenicity and good biosafety by programming neoantigen peptide for nanoassembly with oligonucleotides without the aid of artificial carrier.


Asunto(s)
Antígenos de Neoplasias , Vacunas contra el Cáncer , Péptidos , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/administración & dosificación , Animales , Ratones , Antígenos de Neoplasias/inmunología , Péptidos/inmunología , Péptidos/química , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/inmunología , Oligodesoxirribonucleótidos/química , Células Presentadoras de Antígenos/inmunología , Línea Celular Tumoral , Inmunoterapia/métodos , Humanos , Femenino , Linfocitos T/inmunología , Nanoestructuras/química , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Neoplasias del Colon/tratamiento farmacológico
2.
Front Oncol ; 14: 1336734, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38571505

RESUMEN

Gastric cancer (GC) is a prevalent form of malignancy characterized by significant heterogeneity. The development of a specific prediction model is of utmost importance to improve therapy alternatives. The presence of H. pylori can elicit pyroptosis, a notable carcinogenic process. Furthermore, the administration of chemotherapeutic drugs is often employed as a therapeutic approach to addressing this condition. In the present investigation, it was observed that there were variations in the production of 17 pyroptosis-regulating proteins between stomach tissue with tumor development and GC cells. The predictive relevance of each gene associated with pyroptosis was assessed using the cohort from the cancer genome atlas (TCGA). The least absolute shrinkage and selection operator (LASSO) was utilized to enhance the outcomes of the regression approach. Patients with gastric cancer GC in the cohort from the TCGA were categorized into low-risk or high-risk groups based on their gene expression profiles. Patients with a low risk of gastric cancer had a higher likelihood of survival compared to persons classified as high risk (P<0.0001). A subset of patients diagnosed with GC from a Genes Expression Omnibus (GEO) cohort was stratified according to their overall survival (OS) duration. The statistical analysis revealed a higher significance level (P=0.0063) regarding OS time among low-risk individuals. The study revealed that the GC risk score emerged as a significant independent prognostic factor for OS in patients diagnosed with GC. The results of Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) research revealed that genes associated with a high-risk group had significantly elevated levels of immune system-related activity. Furthermore, it was found that the state of immunity was diminished within this particular group. The relationship between the immune response to cancer and pyroptosis genes is highly interconnected, suggesting that these genes have the potential to serve as prognostic indicators for GC.

3.
Adv Sci (Weinh) ; 11(18): e2309984, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38430531

RESUMEN

The induction of cuproptosis, a recently identified form of copper-dependent immunogenic cell death, is a promising approach for antitumor therapy. However, sufficient accumulation of intracellular copper ions (Cu2+) in tumor cells is essential for inducing cuproptosis. Herein, an intelligent cuproptosis-inducing nanosystem is constructed by encapsulating copper oxide (CuO) nanoparticles with the copper ionophore elesclomol (ES). After uptake by tumor cells, ES@CuO is degraded to release Cu2+ and ES to synergistically trigger cuproptosis, thereby significantly inhibiting the tumor growth of murine B16 melanoma cells. Moreover, ES@CuO further promoted cuproptosis-mediated immune responses and reprogrammed the immunosuppressive tumor microenvironment by increasing the number of tumor-infiltrating lymphocytes and secreted inflammatory cytokines. Additionally, combining ES@CuO with programmed cell death-1 (PD-1) immunotherapy substantially increased the antitumor efficacy in murine melanoma. Overall, the findings of this study can lead to the use of a novel strategy for cuproptosis-mediated antitumor therapy, which may enhance the efficacy of immune checkpoint inhibitor therapy.


Asunto(s)
Cobre , Inmunoterapia , Melanoma Experimental , Animales , Ratones , Inmunoterapia/métodos , Cobre/química , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/inmunología , Modelos Animales de Enfermedad , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ratones Endogámicos C57BL , Línea Celular Tumoral , Clorofilidas , Nanopartículas/química
4.
Cancer Sci ; 114(8): 3396-3410, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37290894

RESUMEN

Gastric cancer (GC) has high rates of morbidity and mortality, and this phenomenon is particularly evident in coastal regions where local dietary habits favor the consumption of pickled foods such as salted fish and vegetables. In addition, the diagnosis rate of GC remains low due to the lack of diagnostic serum biomarkers. Therefore, in this study, we aimed to identify potential serum GC biomarkers for use in clinical practice. To identify candidate biomarkers of GC, 88 serum samples were first screened using a high-throughput protein microarray to measure the levels of 640 proteins. Then, 333 samples were used to validate the potential biomarkers using a custom antibody chip. ELISA, western blot, and immunohistochemistry were then used to verify the expression of the target proteins. Finally, logistic regression was performed to select serum proteins for the diagnostic model. As a result, five specific differentially expressed proteins, TGFß RIII, LAG-3, carboxypeptidase A2, Decorin and ANGPTL3, were found to have the ability to distinguish GC. Logistic regression analysis showed that the combination of carboxypeptidase A2 and TGFß RIII had superior potential for diagnosing GC (area under the ROC curve [AUC] = 0.801). The results suggested that these five proteins alone and the combination of carboxypeptidase A2 and TGFß RIII may be used as serum markers for the diagnosis of GC.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Gástricas , Humanos , Análisis por Matrices de Proteínas , Neoplasias Gástricas/diagnóstico , Carboxipeptidasas A , Detección Precoz del Cáncer , Curva ROC , Proteína 3 Similar a la Angiopoyetina
5.
ACS Sens ; 7(5): 1439-1450, 2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35561250

RESUMEN

SERS-based breath analysis as an emerging technique has attracted increasing attention in cancer screening. Here, eight aldehydes and ketones in the human breath are reported as the VOC biomarkers identified by gas chromatography-mass spectrometry (GC-MS) and applied further for the noninvasive diagnosis of gastric cancer (GC) with a tubular SERS sensor. The tubular SERS sensor is prepared with a glass capillary loaded with ZIF-67-coated silver particles (Ag@ZIF-67), which offers Raman enhancement from the plasmonic nanoparticles and gas enrichment from the metal-organic framework (MOF) shells. The composite materials are modified with 4-aminothiophenol (4-ATP) to capture different aldehyde and ketone compounds. The tubular sensor is served simultaneously as a gas flow channel and a detection chamber, bringing a higher gas capture efficiency than the planar SERS sensor. As a proof-of-concept, the tubular SERS sensor is successfully employed to screen gastric cancer patients with an accuracy of 89.83%, based on the noninvasive, rapid, and easily operated breath analysis. The results demonstrate that the established breath analysis method provides an excellent alternative for the screening of GC and other diseases.


Asunto(s)
Nanopartículas del Metal , Neoplasias Gástricas , Pruebas Respiratorias , Humanos , Plata/química , Espectrometría Raman , Neoplasias Gástricas/diagnóstico
6.
Onco Targets Ther ; 12: 2259-2269, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30988630

RESUMEN

PURPOSE: Gastric carcinoma is the second most frequently diagnosed cancer and leading cause of cancer death in China. As a new generation of cancer therapeutic drug, CL-6, a curcumin derivative, shows better bioavailability than curcumin, which has shown anticancer effects in gastric cancer (GC). However, whether CL-6 shows similar activities in GC has not been examined. MATERIALS AND METHODS: Cell proliferation assay, colony-forming assay, flow cytometric analysis, wound healing assay, and Transwell invasion assay were performed to examine the effects of CL-6 on proliferation, apoptosis, migration, and invasion on human AGS and MGC-803 cell lines. Western blot was used to evaluate protein levels of Bax, Bcl-2, YAP, p-YAP, and Lats, and gene expression was measured by real-time quantitative PCR (RT-qPCR). RESULTS: CL-6 dose dependently reduced proliferation, increased apoptosis, and inhibited the migration and invasion abilities of AGS and MGC-803 cells. CL-6 also increased levels of pro-apoptotic protein Bax, decreased levels of antiapoptotic protein Bcl-2, and increased the Bax/Bcl-2 ratio. CL-6 treatment also inhibited YAP and YAP protein and mRNA expression, while it induced the expression of Lats and p-YAP (Ser127). CONCLUSION: CL-6 induces apoptosis of GC cells by activating the Hippo-YAP signaling pathway. These results indicate the therapeutic potential of the novel curcumin derivative CL-6 in GC.

7.
Mol Ther ; 27(5): 1039-1050, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-30852137

RESUMEN

Histone deacetylase inhibitors (HDACis) are the recommended treatment for many solid tumors; however, resistance is a major clinical obstacle for their efficacy. High levels of the transcription factor nuclear factor erythroid 2 like-2 (Nrf2) in cancer cells suggest a vital role in chemoresistance, and regulation of autophagy is one mechanism by which Nrf2 mediates chemoresistance. Although the molecular mechanisms underlying this activity are unclear, understanding them may ultimately improve therapeutic outcomes following HDACi treatment. In this study, we found that HDACi treatment increased Nrf2 mRNA and protein levels and enhanced Nrf2 transcriptional activity. Conversely, Nrf2 knockdown or inhibition blocked HDACi-induced autophagy. In addition, a microRNA (miRNA) array identified upregulation of miR-129-3p in response to Nrf2 overexpression. Chromatin immunoprecipitation assays confirmed miR-129-3p to be a direct Nrf2 target. RepTar and RNAhybrid databases indicated mammalian target of rapamycin (mTOR) as a potential miR-129-3p target, which we experimentally confirmed. Finally, Nrf2 inhibition or miR-129-3p in combination with HDACis increased cell death in vitro and in vivo. Collectively, these results demonstrated that Nrf2 regulates mTOR during HDACi-induced autophagy through miRNA-129-3p and inhibition of this pathway could enhance HDACi-mediated cell death.


Asunto(s)
MicroARNs/genética , Factor 2 Relacionado con NF-E2/genética , Neoplasias/tratamiento farmacológico , Serina-Treonina Quinasas TOR/genética , Animales , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Redes Reguladoras de Genes/genética , Xenoinjertos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ratones , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Neoplasias/genética , Neoplasias/patología
8.
Life Sci ; 201: 141-149, 2018 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-29601890

RESUMEN

AIMS: Gastrointestinal cancers are a kind of deadly malignancy afflicting close to a million peoples worldwide. 5-Fluorouracil (5-Fu) is a main chemotherapeutic agent for cancer treatment. However, prolonged exposure of 5-Fu to cancer cells may cause chemoresistance and decrease the therapeutic potential of 5-Fu. MAIN METHODS: Replication protein A (RPA) is a component of the origin recognition complex. In our study, we explored the role of RPA1 in hepatocellular carcinoma cell SMMC-7721, gastric cancer cell SGC-7901 and colorectal cancer HT-29 via lentiviral particles infection. Flow cytometry assay was used to examine the effect of RPA1 on cell proliferation, cell cycle and apoptosis. Western blot was employed to determine the role of RPA1 on Caspase 3 expression. KEY FINDINGS: Immunohistochemstry results showed that RPA1 was highly expressed in colorectal cancer tissues. Only 5-Fu or the knockdown of RPA1 suppressed cell clone formation, induced cell cycle arrest at the G1 phase and promoted cell apoptosis by regulating the protein level of Caspase 3. And the combination of the application of 5-Fu and RPA1 silencing significantly enhanced the above effects. SIGNIFICANCE: RPA1 serves as an oncogene during gastrointestinal cancers progression. These studies reveal a new target for gastrointestinal cancers therapy, and the combination of 5-Fu and silencing of RPA1 provides a new attractive therapeutic measure for gastrointestinal cancers.


Asunto(s)
Apoptosis/genética , Caspasa 3/biosíntesis , Silenciador del Gen , Proteína de Replicación A/genética , Caspasa 3/genética , Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/patología , Células HT29 , Humanos , Ensayo de Tumor de Célula Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA