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

Banco de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
BMC Med ; 20(1): 376, 2022 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-36310169

RESUMEN

BACKGROUND: The combination of oncolytic viruses (OVs) with immune checkpoint blockades is a research hotspot and has shown good efficacy. Here, we present the first attempt to combine oncolytic herpes simplex virus 2 (OH2) with an anti-SIRPα antibody as an antitumour treatment. Our results provide unique insight into the combination of innate immunity with OV. METHODS: We verified the polarization and activation of OH2 in RAW264.7 cells in vitro. Subsequently, we evaluated the antitumour ability of OH2 and anti-SIRPα combined therapy in a tumour-bearing mouse model. RNA-seq and Single-cell RNA-seq were used to characterize the changes in the tumour microenvironment. RESULTS: The OH2 lysates effectively stimulated RAW264.7 cells to polarize towards the M1 but not the M2 phenotype and activated the function of the M1 phenotype in vitro. In the macrophage clearance experiment, OH2 therapy induced polarization of M1 macrophages and participated in the antitumour immune response in a tumour-bearing mouse model. Treatment with a combination of OH2 and anti-SIRPα effectively inhibited tumour growth and significantly prolonged the survival time of the mice, and this result was more obvious in the mouse model with a larger tumour volume at the beginning of the treatment. These results suggest that combination therapy can more profoundly reshape the TME and activate stronger innate and adaptive immune responses. CONCLUSIONS: Our data support the feasibility of oncolytic virus therapy in combination with anti-SIRPα antibodies and suggest a new strategy for oncolytic virus therapy.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Ratones , Animales , Virus Oncolíticos/genética , Microambiente Tumoral , Viroterapia Oncolítica/métodos , Neoplasias/terapia , Inmunidad Innata , Modelos Animales de Enfermedad
2.
Cancer Innov ; 3(1): e101, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38948532

RESUMEN

In recent years, the three-dimensional (3D) culture system has emerged as a promising preclinical model for tumor research owing to its ability to replicate the tissue structure and molecular characteristics of solid tumors in vivo. This system offers several advantages, including high throughput, efficiency, and retention of tumor heterogeneity. Traditional Matrigel-submerged organoid cultures primarily support the long-term proliferation of epithelial cells. One solution for the exploration of the tumor microenvironment is a reconstitution approach involving the introduction of exogenous cell types, either in dual, triple or even multiple combinations. Another solution is a holistic approach including patient-derived tumor fragments, air-liquid interface, suspension 3D culture, and microfluidic tumor-on-chip models. Organoid co-culture models have also gained popularity for studying the tumor microenvironment, evaluating tumor immunotherapy, identifying predictive biomarkers, screening for effective drugs, and modeling infections. By leveraging these 3D culture systems, it is hoped to advance the clinical application of therapeutic approaches and improve patient outcomes.

3.
Cancer Lett ; 588: 216757, 2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38417668

RESUMEN

Circulating tumor cell (CTC) clusters exhibit significantly higher metastatic potential compared to single CTCs. However, the underlying mechanism behind this phenomenon remains unclear, and the role of posttranscriptional RNA regulation in CTC clusters has not been explored. Here, we conducted a comparative analysis of alternative splicing (AS) and alternative polyadenylation (APA) profiles between single CTCs and CTC clusters. We identified 994 and 836 AS events in single CTCs and CTC clusters, respectively, with ∼20% of AS events showing differential regulation between the two cell types. A key event in this differential splicing was observed in SRSF6, which disrupted AS profiles and contributed to the increased malignancy of CTC clusters. Regarding APA, we found a global lengthening of 3' UTRs in CTC clusters compared to single CTCs. This alteration was primarily governed by 14 core APA factors, particularly PPP1CA. The modified APA profiles facilitated the cell cycle progression of CTC clusters and indicated their reduced susceptibility to oxidative stress. Further investigation revealed that the proportion of H2AFY mRNA with long 3' UTR instead of short 3' UTR was higher in CTC clusters than single CTCs. The AU-rich elements (AREs) within the long 3' UTR of H2AFY mRNA enhance mRNA stability and translation activity, resulting in promoting cell proliferation and invasion, which potentially facilitate the establishment and rapid formation of metastatic tumors mediated by CTC clusters. These findings provide new insights into the mechanisms driving CTC cluster metastasis.


Asunto(s)
Células Neoplásicas Circulantes , Humanos , Células Neoplásicas Circulantes/patología , Regiones no Traducidas 3' , Poliadenilación , Empalme del ARN , Análisis por Conglomerados , Metástasis de la Neoplasia , Factores de Empalme Serina-Arginina/metabolismo , Fosfoproteínas/metabolismo
4.
Cell Death Discov ; 8(1): 6, 2022 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013129

RESUMEN

Signet ring cell carcinoma (SRCC) has specific oncogenesis and phenotypic and treatment resistance heterogeneity. Systemic therapies are often ineffective, and predictive biomarkers to guide treatment are urgently needed. Tumor organoids have recently emerged as an ideal model for drug testing and screening. Here, we report gastric organoids established from tumor tissues comprising four SRCCs and eight non-SRCCs. Tumor organoids demonstrated different growth characteristics and morphologies. Changes in the original tumor genome were maintained during long-term culture from whole-exome sequencing (WES) analysis. Immunohistochemistry and H&E staining showed that the tissue characteristics of the primary tumor could be recapitulated. In addition, organoid lines successfully formed tumors in immunodeficient mice and maintained tumorigenic character. Different responses to 5-fluorouracil, oxaliplatin, docetaxel and irinotecan treatment were observed in SRCC and non-SRCC organoids. These results demonstrate that gastric organoid drug models, including SRCC, were highly similar to the original tumors in phenotypic and genotypic profiling and could be as living biomarkers for drug response testing.

5.
Transl Androl Urol ; 11(6): 803-813, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35812202

RESUMEN

Background: Whether circulating tumor cells (CTCs) with prostate-specific membrane antigen (PSMA) high expression was related to the metastatic progress in prostate cancer (PCa) remains explored. This study aimed to provide evidence to elucidate this relationship via the telomerase reverse transcriptase (TERT)-based CTC detection method. Methods: A total of 71 patients were enrolled and divided into the local PCa group (n=44) and metastatic PCa group (n=27). TERT-based CTC detection (TBCD) was used to detect CTCs. CTCs single-cell sequencing data were analyzed using gene ontology (GO) functional classification and enrichment. Results: The mean 'TERT+ CTCs' number was 6.11±9.63 in the metastatic group and 4.09±3.41 in the local group. GO enrichment analysis for 77 prostate CTCs single-cell sequencing confirmed that proliferation-related terms were enriched in the PSMA-high expression group, and 27 metastasis-related gene panels also had high expression in this group. Then, PSMA antibody was applied to mark the 'TERT+ CTCs'. The proportion of patients with 'TERT+ PSMA+ CTCs' was positively associated with the Gleason score. Furthermore, the proportion of 'TERT+ PSMA+ CTCs' patients was 48.15% in the metastatic group, significantly higher than 22.72% in the local group. Conclusions: This study suggested that TERT positive CTCs with high PSMA expression were associated with the PCa metastatic progress.

6.
Theranostics ; 12(17): 7371-7389, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36438484

RESUMEN

Rationale: Oncolytic virus (OV) therapy as a cancer therapy that improves immune status makes it a favorable candidate for optimizing immunotherapy strategies. Existing studies have focused on characterizing the disturbance of the tumor microenvironment (TME) by OV therapy. However, the changes in systemic immunity induced by OV were largely ignored, which would prevent the further understanding and optimization of oncolytic viruses. Methods: The HSV-2-based oncolytic virus OH2 was used to treat tumor-bearing mouse models. The peripheral blood samples were then collected for single-cell RNA sequencing (scRNA-seq). The scRNA-seq data were analyzed using Cell Ranger, Seurat, and other bioinformatics tools. Key findings were further validated by ELISA, immunohistochemistry, flow cytometry, in vivo experiments, and clinical samples. Results: Our data showed that OH2 therapy effectively activated systemic immunity and induced a sustained anti-tumor immune response. One major impact of OH2 on systemic immunity was to boost Ccl5 production, which correlated with clinical response. Besides, the cytotoxic ability of peripheral cytotoxic Cd8+ T cells and mature NK cells was elevated by OH2. Further analysis revealed that the interaction of monocytes with T cells and NK cells was critical for systemic immune remodeling and activation. We also found that systemic immune responses induced by OH2 could effectively reshape the microenvironment of distant tumor lesions and inhibit their progression. Conclusions: This study is the first to comprehensively characterize the effects of OV therapy on systemic immunity, which not only sheds new light on the anti-tumor mechanisms of OH2, but also contributes to the establishment of companion diagnostics for OH2 treatment and the improvement of oncolytic therapy strategies.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Ratones , Animales , Transcriptoma , Virus Oncolíticos/genética , Inmunoterapia , Microambiente Tumoral , Neoplasias/patología
7.
Infect Drug Resist ; 13: 3679-3687, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33116687

RESUMEN

This study analyzed the barriers of patient access to affordable MDR-TB medication in China and the reasons behind, and proposed strategies towards removing the barriers based on literature review and key informant interviews. Reasons behind the high financial burden of MDR-TB patients in China are the lack of a coordinated and multi-sourced financing model to secure patients' access to the expensive novel medicines, and the absence of the safety-net for the patients with low ability to pay the costs. Appropriate health insurance benefit packages and provider payment mechanisms, supportive legal framework, coordinated policies as well as incentives for off-label use of evidence-based repurposed medicines are missing. The observations identified key intervention areas including continued efforts to make the novel effective medicines affordable and to strengthen the legislative protection for off-label use of evidence-based medicines; increase incentives for pharmaceutical companies to expand indications of established medicines based on the evidence; implement public initiatives to support the use of repurposed medicines for diseases with major public health significance, and scale up good practices from local pilots to create a coordinated multi-sourced financing model. A comprehensive approach to address the barriers in the full treatment course of MDR-TB and a safety-net for low-ability-to-pay patients are also critical to secure universal access to affordable MDR-TB medication.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA