Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Discov Oncol ; 15(1): 124, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38639872

RESUMO

The role of SLC35A2 in breast cancer remains poorly understood, with limited available information on its significance. This study aimed to investigate the expression of SLC35A2 and clinicopathological variables in breast cancer patients. Immunohistochemical analysis of SLC35A2 protein was conductedon 40 adjacent non-neoplastic tissues and 320 breast cancer tissues. The study also assesed the association between SLC35A2 expression and breast cancer clinicopathological features of breast cancer, as well as its impact on overall survival. In comparison to adjacent non-neoplastic tissues, a significantly higher expression of SLC35A2 was observed in breast cancer tissues (P = 0.020), and this expression was found to be independently correlated with HER2 positivity (P = 0.001). Survival analysis indicated that patients with low SLC35A2 expression had a more favorable prognosis in HER2-positive subtype breast cancer (P = 0.017). These results suggest that SLC35A2 is overexpressed in breast cancer tissues compared to adjacent non-neoplastic tissues and may serve as a potential prognostic marker for HER2-positive subtype breast cancer. Furthermore, breast cancer patients with the HER2 positive subtype who exhibited decreased levels of SLC35A2 expression demonstrated improved long-term prognostic outcomes.

2.
Oncol Lett ; 26(4): 418, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37664666

RESUMO

Malignant ascites (MA) is a common manifestation of advanced gastric cancer (GC) with peritoneal metastasis (PM), which usually indicates a poor prognosis. The present study aimed to explore the effects of MA, a unique microenvironment of PM, on the proliferation of cancer cells and investigate the underlying mechanisms. Ex vivo experiments demonstrated that GC cells treated with MA exhibited enhanced proliferation. RNA sequencing indicated that asparagine synthetase (ASNS) was one of the differentially expressed genes in GC cells following incubation with MAs. Furthermore, the present study suggested that MA induced an upregulation of ASNS expression and the stimulatory effect of MA on cancer cell proliferation was alleviated upon ASNS downregulation. Activating transcription factor 4 (ATF4), a pivotal transcription factor regulating ASNS, was upregulated when cells were treated with MA supernatant. After ATF4 knockdown, the proliferation of MA-treated GC cells and the expression of ASNS decreased. In addition, the decline in the proliferation of the ATF4-downregulated AGS GC cell line was rescued by ASNS upregulation. The findings indicated that MA could promote the proliferation of GC cells via activation of the ATF4-ASNS axis. Hence, it may be a potential target for treating GC with PM and MA.

3.
Ther Adv Med Oncol ; 15: 17588359231187205, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37484525

RESUMO

Immune checkpoint inhibitors (ICIs) targeting programmed cell death 1, programmed cell death ligand 1, and cytotoxic T lymphocyte-associated antigen-4 have shown significantly durable clinical benefits and tolerable toxicities and have improved the survival of patients with various types of cancer. Since 2018, the National Medical Products Administration of China has approved 17 ICIs as the standard treatment for certain advanced or metastatic solid tumors. As ICIs represent a broad-spectrum antitumor strategy, the populations eligible for cancer immunotherapy are rapidly expanding. However, the clinical applications of ICIs in cancer patient populations with special issues, a term that refers to complex subgroups of patients with comorbidities, special clinical conditions, or concomitant medications who are routinely excluded from prospective clinical trials of ICIs or are underrepresented in these trials, represent a great real-world challenge. Although the Chinese Society of Clinical Oncology (CSCO) has provided recommendations for screening before the use of ICIs in special populations, the recommendations for full-course management remain insufficient. The CSCO Expert Committee on Immunotherapy organized leading medical oncology and multidisciplinary experts to develop a consensus that will serve as an important reference for clinicians to guide the proper application of ICIs in special patient populations. This article is a translation of a study first published in Chinese in The Chinese Clinical Oncology (ISSN 1009-0460, CN 32-1577/R) in May 2022 (27(5):442-454). The publisher of the original paper has provided written confirmation of permission to publish this translation in Therapeutic Advances in Medical Oncology.

4.
Sci Rep ; 13(1): 11125, 2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37429893

RESUMO

Gastric cancer remains one of the most prevalent tumors worldwide and peritoneal metastasis is responsible for approximately 60% of death in advanced gastric cancer patients. However, the underlying mechanism of peritoneal metastasis is poorly understood. We have established organoids derived from malignant ascites (MA) of gastric cancer patients and noticed that MA supernatant could strongly increase the colony formation of organoids. Thus, we realized the interaction between exfoliated cancer cells (ECCs) and liquid tumor microenvironment contributes to peritoneal metastasis. Further, we designed a medium component control test which proved that exosomes derived from MA could not enhance the growth of organoids. Using Immunofluorescence and confocal imaging as well as dual-luciferase reporter assay, our data showed WNT signaling pathway was upregulated by high concentrations of WNT ligands (wnt3a and wnt5a), which was verified by ELISA. Besides, suppressing WNT signaling pathway diminished the growth promoting function of MA supernatant. This result implicated WNT signaling pathway as a potential therapeutic target for peritoneal metastasis of gastric cancer.


Assuntos
Neoplasias Peritoneais , Neoplasias Gástricas , Humanos , Via de Sinalização Wnt , Microambiente Tumoral , Peritônio
5.
DNA Cell Biol ; 42(7): 372-389, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37285280

RESUMO

Infiltrated immune cells are an important constitute of tumor microenvironment, which exert complex effects on gastric cancer (GC) pathogenesis and progression. By using weighted gene co-expression network analysis, integrating the data from The Cancer Genome Atlas-stomach adenocarcinoma and GSE62254, we identify Aldo-Keto Reductase Family 1 Member B (AKR1B1) as a hub gene for immune regulation in GC. Notably, AKR1B1 is associated with higher immune infiltration and worse histologic grade of GC. In addition, AKR1B1 is an independent factor for predicting the survival rate of GC patients. In vitro experiments further demonstrated that AKR1B1-overexpressed THP-1-derived macrophages promoted the proliferation and migration of GC cells. Taken together, AKR1B1 plays an important role in GC progression by regulating immune microenvironment, which could be a biomarker for predicting GC prognosis as well as a potential therapeutic target for GC treatment.


Assuntos
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Microambiente Tumoral/genética , Aldeído Redutase/genética
6.
Am J Transl Res ; 15(4): 2568-2584, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37193151

RESUMO

BACKGROUND: Recent researches found that mitochondrial functions were substantially involved in tumor progression, whereas the particular mechanism is unrecognized. Coiled-Coil Domain-Containing Protein 58 (CCDC58), one of the mitochondrial matrix import factors, acts as a novel regulator or stabilizer involved in mitochondrial protein import machinery. Whether and how an up-regulation of CCDC58 causes poor prognosis of patients in Hepatocellular Carcinoma (HCC) still required further researches. METHODS: Tumor immune estimation resource (TIMER), Hepatocellular Carcinoma Database (HCCDB) and UALCAN databases were utilized to explore the expression level in diverse types of tumors compared with normal tissues. The prognostic potential of CCDC58 mRNA was evaluated via the Kaplan-Meier plotter, Gene Expression Profiling Interactive Analysis (GEPIA) and the Human Protein Atlas (HPA) databases. Corresponding clinicopathological factors were analyzed in Kaplan-Meier plotter. According to the median of mRNA expression levels of CCDC58, we divided The Cancer Genome Atlas (TCGA) data of HCC patients into two groups, highly expressed one and lowly expressed one, so as to perform the enrichment analyses of Gene Oncology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Protein-Protein Interaction (PPI) Network was constructed by STRING site and the co-expressed genes were functionally enriched. Immunohistochemistry was adopted to detect protein expression of CCDC58 in HCC patients. RESULTS: This study indicated that CCDC58 protein expression level was obviously higher in HCC than that in paired paracancerous tissues. The up-regulated CCDC58 mRNA is prone to poor prognosis of patients in HCC through various indexes, such as overall survival (OS), disease-free survival (DFS), disease-specific survival (DSS), relapse-free survival (RFS) and progression-free survival (PFS). Additionally, univariate and multivariate Cox regression analyses suggested that CCDC58 could be viewed as an independent risk factor for HCC patients. The expression of CCDC58 is associated with 28 GO terms related to mitochondria and 5 KEGG pathways including oxidative phosphorylation. The PPI network revealed 10 interactive proteins about constituent components of mitochondria. CONCLUSIONS: These findings demonstrated CCDC58 to be a potential diagnostic and prognostic biomarker in HCC and correlated with mitochondria acting on tumor biosynthesis and energy production. It is reliable for CCDC58 to be targeted to design novel treatments for HCC patients.

7.
Am J Transl Res ; 15(1): 316-323, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36777837

RESUMO

The presence of peritoneal metastasis in patients with pancreatic cancer is associated with poor prognosis. Chemotherapy and radiotherapy may result in poor prognosis in patients with pancreatic cancer. However, immunotherapy improves prognosis even at an advanced stage of the disease. The present study reported a case of a combined therapy of autologous ex vivo expanded natural killer (NK) cells and programmed cell death 1 (PD-1) inhibitor in a patient with pancreatic cancer and peritoneal metastasis. The NK cells were expanded ex vivo and intravenously injected. This was followed by intravenous administration of two dosages of PD-1 inhibitor. Computed tomography and magnetic resonance imaging were performed to assess the size of tumor before and after the combined therapy. In addition, the blood sample and ascites were collected and analyzed before and after the combined therapy. Flow cytometry was carried out to measure the subsets of T cells and macrophages in the collected ascites. Meanwhile, the levels of cytokines in the ascites were quantified through enzyme-linked immunosorbent assay, and Luminex assays were conducted on the supernatant. It was revealed that after the combined therapy, cancer cells disappeared in the ascites, and the T cells were activated, which could be confirmed by the decreased levels of PD-1 and T cell immunoglobulin and mucin domain-containing protein 3. Also, the functioning of macrophages was improved, as shown by the increased level of CD86 and the reduced levels of CD206 and HLA-DR. Notably, the levels of cytokines (transforming growth factor-ß, vascular endothelial growth factor, and interleukin-10) in ascites were significantly upregulated after the combined therapy. In conclusion, it was evident that NK cells combined with PD-1 inhibitor improved the immune microenvironment of carcinomatosis in the peritoneal cavity. Therefore, the combined therapy may be beneficial for suppressing pancreatic cancer and the presence of metastases in the peritoneal cavity. However, there is a need for additional randomized studies to confirm the efficacy of combined therapy.

8.
Eur J Pharm Sci ; 182: 106373, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36623698

RESUMO

OBJECTIVE: The aim of this study was to examine the pharmacokinetics, bioequivalence, and safety of two tablet formulations of capecitabine 500 mg in Chinese patients with breast, colorectal or gastric cancer under fed condition. METHODS: A multicentric, randomized, open-label, single-dose, two-period, two-way crossover trial was conducted by randomizing a single oral dose of test (T) or reference (R, Xeloda®) capecitabine (500 mg) to patients of either sex with colon, colorectal or breast cancer under fed condition (high-fat and high-calorie diet). Pharmacokinetic parameters were calculated using non-compartmental methods. Patients were monitored for safety and tolerability throughout the study. RESULTS: 74 subjects were randomly enrolled. The T/R geometric mean ratios (GMRs) and 90% confidence intervals (CIs) for Cmax, AUC0-t and AUC0-∞ of capecitabine were 96.60% (85.87-108.67%), 99.07% (95.40-102.89%), 99.17% (95.29-103.21%), respectively. All 90% CIs fell within the bioequivalence acceptance range of 80.00-125.00%. The common adverse events (AEs) included clinically significant laboratory abnormalities and gastrointestinal diseases. There were no serious adverse events (SAEs) or deaths during the study. No subject withdrew from the study due to AEs. CONCLUSION: Single oral intake of test and the reference capecitabine tablets were bioequivalent under fed condition and had similar favourable safety profiles in Chinese patients with breast, colorectal or gastric cancer. TRIAL REGISTRATION: chinadrugtrials.org.cn (CTR20182110).


Assuntos
Neoplasias Colorretais , Neoplasias Gástricas , Humanos , Área Sob a Curva , Capecitabina/efeitos adversos , China , Estudos Cross-Over , População do Leste Asiático , Jejum , Neoplasias Gástricas/tratamento farmacológico , Comprimidos , Equivalência Terapêutica
9.
J Cancer Res Clin Oncol ; 149(7): 3803-3810, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35987927

RESUMO

OBJECTIVE: To establish an in vitro study model of gastric cancer, gastric cancer organoid culture system, by biopsy under gastroscope, and to explore and analyze the related factors affecting the success rate of culture, to provide a better in vitro model for the study of gastric cancer. METHODS: Twenty-six patients with advanced gastric cancer were collected. Organoids were cultured by biopsy under gastroscope. Paraffin sections were made and HE staining was used to compare the consistency of gastroscopic pathological morphology and organoids. To explore the influencing factors of cultivating gastric cancer organoids in combination with clinical data. RESULTS: A total of 26 cases were collected by gastroscopy, and 12 cases of gastric cancer organoids were successfully cultured after identification, with a success rate of about 48%. Its histopathological morphology was highly consistent with that of gastric cancer. According to the pathological type, 21 cases were poorly differentiated adenocarcinoma and 12 cases were successful. Four cases of signet ring cell carcinoma failed. According to the location of the lesion, the success rate of sampling and culture of gastric antrum was significantly lower, which may be related to antral edema and anatomical characteristics of gastric antrum. Some of the failed cases are related to the quality of sampling, technique and contamination of tissue cells. CONCLUSIONS: We have successfully established gastric cancer organoids through endoscopic biopsy, and analyzed the factors affecting the success rate of culture from various angles, to improve and enhance the organoid culture technology and provide a better platform for tumor research.


Assuntos
Adenocarcinoma , Carcinoma de Células em Anel de Sinete , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patologia , Adenocarcinoma/patologia , Biópsia , Organoides/patologia
10.
Front Oncol ; 12: 876899, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36330490

RESUMO

Background and purpose: The objective of this study was to investigate the pharmacokinetics, safety, and antitumor activity of apatinib, a vascular endothelial growth factor receptor 2 inhibitor, in advanced gastric adenocarcinoma or gastroesophageal junction adenocarcinoma and evaluate the effect of dose titration on dosage optimization for individual patients. Methods: Patient with advanced gastric adenocarcinoma progressed after at least one line of chemotherapy were enrolled. Apatinib was given orally once daily starting at 500 mg for 14 days, then up-titrated to 750 mg for 14 days, and then proceeded to a maximum dose of 850 mg. Dose up-titration determination was based on toxicity. The 28-day treatment cycles continued until disease progression, intolerable toxicities, withdrawal of consent, or investigator' decision. Results: A total of 60 patients were enrolled, with 17, 18, and 25 patients receiving a maximum dose of 500 mg, 750 mg, and 850 mg, respectively. The pharmacokinetic parameters varied considerably, with the interpatient coefficient of variation for steady state areas under the plasma concentration time curve (AUCss) and the mean maximum concentration of both > 50%. During 500 mg and 750 mg dosing stage, drug exposures in patients with a maximum dosage of 850 mg were lower than in those not titrated to 850 mg. Patients with total gastrectomy exhibited significantly lower AUCss than patients with partial or no gastrectomy (p = 0.004 and 0.032, respectively). Toxicities were tolerable, and disease control rate was 39.5% (95% CI 25.0%-55.6%). Conclusions: Apatinib dose titration based on toxicity could be used in clinical practice to provide optimal dosage for individual patients. Clinical Trial registration: https://clinicaltrials.gov/ct2/show/NCT02764268?term=NCT02764268&draw=2&rank=1, NCT02764268.

11.
Front Oncol ; 12: 1054598, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36439457

RESUMO

In recent years, numerous studies have demonstrated that the tumor immune microenvironment (TIME) is capable of regulating the growth of tumors, and tumor-infiltrating immune cells in the TIME can affect the prognosis and treatment responses of patients. Consequently, therapies targeting these immune cells have emerged as important antitumor treatments. As a crucial componet of the perioperative treatment of malignant tumors, neoadjuvant chemotherapy (NACT) can improve the surgical resection rate and prognosis of patients and is a suitable clinical model to evaluate the effect of chemotherapy on the TIME. To provide a rationale for developing valid combinational therapies, this review summarizes the impact of NACT on the TIME, the relationship between tumor-infiltrating immune cells and treatment responses of patients, and the prognostic value of these infiltrating immune cells.

12.
Front Immunol ; 13: 984149, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36300121

RESUMO

Background: N7-methylguanosine (m7G), one of the most common post-transcriptional modifications, can be present in tRNA, mRNA, and miRNA to mediate the progression of various tumors. However, the possible role of m7G in gastric cancer (GC) is still unknown. Materials and Methods: In this study, SNVs (single nucleotide variations), CNVs (copy number variations), and methylation of m7G-related genes (m7GRGs) were analyzed. The relationship between them and the expression of m7GRGs and prognosis of GC patients was explored. Based on 13 prognostic-related m7GRGs, 567 GC samples were classified into three subtypes using the ConsensusClusterPlus package. we compared survival status, clinical traits, immune cell infiltration, immune checkpoints, tumor microenvironment (TME), tumor immune dysfunction and exclusion (TIDE), and potential biological pathways among the three subtypes. Then, patients were again grouped into different genetic subtypes based on the DEGs among the three subtypes. In addition, a prognostic m7GRG_Score was constructed using five risk genes applicable to patients of any age, gender and stage. We also assessed tumor mutational burden (TMB), microsatellite instability (MSI), cancer stem cell (CSC) index, sensitivity of antineoplastic drugs, efficacy of anti-PD-1 and anti-CTLA4 immunotherapy between high and low m7GRG_Score groups. Finally, we established a nomogram based on m7GRG_Score and tumor stage to enhance the clinical application of the model. miRNAs and lncRNAs that could regulate expression of risk genes were searched. Results: SNVs, CNVs, and methylation of m7GRGs were associated with m7GRGs expression. However, they did not significantly affect the survival of GC patients. Our results also confirmed that patients in subtypes B and C and low m7GRG_Score groups had longer survival time, better clinical stage, more immune cell infiltration, fewer immune escape and dysfunction compared to subtype A and high m7GRG_Score groups. A low m7GRG_score was featured with increased microsatellite instability-high (MSI-H), TMB, and efficacy of immunotherapy. Conclusion: The m7GRG_Score model may become a beneficial tool for predicting prognosis and guiding personalized treatment in GC patients. These findings will improve our knowledge of m7G in GC and provide new methods for more effective treatment strategies.


Assuntos
MicroRNAs , RNA Longo não Codificante , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patologia , Prognóstico , Instabilidade de Microssatélites , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Variações do Número de Cópias de DNA , Biomarcadores Tumorais/genética , MicroRNAs/genética , RNA Mensageiro , Nucleotídeos , Microambiente Tumoral/genética
13.
Front Oncol ; 12: 911410, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35965509

RESUMO

As a significant public health problem with high morbidity and mortality worldwide, tumor is one of the major diseases endangering human life. Moreover, metastasis is the most important contributor to the death of tumor patients. Epithelial-mesenchymal transition (EMT) is an essential biological process in developing primary tumors to metastasis. It underlies tumor progression and metastasis by inducing a series of alterations in tumor cells that confer the ability to move and migrate. Tumor-associated macrophages (TAMs) are one of the primary infiltrating immune cells in the tumor microenvironment, and they play an indispensable role in the EMT process of tumor cells by interacting with tumor cells. With the increasing clarity of the relationship between TAMs and EMT and tumor metastasis, targeting TAMs and EMT processes is emerging as a promising target for developing new cancer therapies. Therefore, this paper reviews the recent research progress of tumor-associated macrophages in tumor epithelial-mesenchymal transition and briefly discusses the current anti-tumor therapies targeting TAMs and EMT processes.

14.
Oncol Lett ; 22(3): 647, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34386069

RESUMO

Pancreatic cancer, a highly malignant disease, is characterized by rapid progression and early metastasis. Although the integrative treatment of pancreatic cancer has made great progress, the prognosis of patients with advanced pancreatic cancer remains extremely poor. In recent years, with the advancements in tumor immunology, immunotherapy has become a promising remedy for pancreatic cancer. Natural killer (NK) cells are the key lymphocytes in the innate immune system. NK cell function does not require antigen pre-sensitization and is not major histocompatibility complex restricted. By targeting tumors or virus-infected cells, the cells play a key role in immune surveillance. Although several questions about NK cells in pancreatic cancer still need to be further studied, there are extensive theories supporting the clinical application prospects of NK cell immunotherapy in pancreatic cancer. Since very few studies have evaluated the role of NK cells in pancreatic cancer, this review provides a comprehensive update of the role of NK cells in pancreatic cancer immunotherapy.

15.
Exp Ther Med ; 21(4): 340, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33732313

RESUMO

The chimeric antigen receptor (CAR) is an artificially modified fusion protein consisting of an extracellular antigen-binding domain, transmembrane domain and intracellular signalling domain. CAR-T therapy has demonstrated remarkable clinical efficacy in hematological malignancies. However, cytokine release syndrome and other side effects have hindered its application in solid tumors. CAR-natural killer (NK) cells have attracted broad attention due to their safety in clinical applications, their mechanism in recognising cancer cells and the abundance of its clinical specimens. Preclinical and clinical trials of human primary NK cells and NK-92 cell lines demonstrated that CAR-NK cells are able to fight haematological malignancies and solid tumors. However, the implication of CAR-NK cell therapy also has certain challenges, including the expansion and activation of primary NK cells in vitro, selection of CAR targets, survival time of CAR-NK cells in vivo, storage and transportation of NK cells, and efficiency of NK cell transduction. This review focuses on the latest progress of CAR-NK cells in the treatment of solid tumors.

16.
Mol Med Rep ; 23(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33760113

RESUMO

Pancreatic mucinous cystadenocarcinoma (MCC) is a rare malignant tumor, with a limited number of studies. The present study aimed to investigate the function and mechanism of microRNA (miR)­224­5p on proliferation, migration and invasion of MCC of the pancreas. Reverse transcription­quantitative PCR was used to explorethe expression of miR­224­5p and the PTEN gene. MTT, wound healing, Transwell and tumorigenesis assays were conducted to investigate the proliferation, migration and invasion of MCC1 cells in vitro and in vivo. Western blot analysis was employed to test the protein expression of PTEN. The target gene of miR­224­5p was assessed and verified by luciferase assay. miR­224­5p expression was notably higher, while PTEN expression was lower, in MCC1 cells compared with normal tissues and cells. Overexpression of miR­224­5p promoted the proliferation, migration and invasion of MCC and knockdown of miR­224­5p inhibited these functions. Bioinformatics analysis and luciferase assay indicated that PTEN was the direct target gene of miR­224­5p. The negative correlation between miR­224­5p and PTEN was confirmed both in vitro and in vivo. PTEN reversed the effects of miR­224­5p on proliferation, migration and invasion of MCC1 cells. The present study revealed for the first time, to the best of the authors' knowledge, that miR­224­5p was highly expressed and served an oncogenic role in MCC. miR­224­5p not only regulated the proliferation, migration and invasion of pancreatic MCC but may also be a potential therapeutic target for MCC.


Assuntos
Cistadenocarcinoma Mucinoso/genética , MicroRNAs/genética , PTEN Fosfo-Hidrolase/genética , Neoplasias Pancreáticas/genética , Idoso , Animais , Apoptose , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Cistadenocarcinoma Mucinoso/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Xenoenxertos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Neoplasias Pancreáticas/patologia
17.
Sci Rep ; 11(1): 1141, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33441952

RESUMO

Gastric cancer (GC) is a leading cause of cancer-induced mortality, with poor prognosis with metastasis. The mechanism of gastric carcinoma lymph node metastasis remains unknown due to traditional bulk-leveled approaches masking the roles of subpopulations. To answer questions concerning metastasis from the gastric carcinoma intratumoural perspective, we performed single-cell level analysis on three gastric cancer patients with primary cancer and paired metastatic lymph node cancer tissues using single-cell RNA-seq (scRNA-seq). The results showed distinct carcinoma profiles from each patient, and diverse microenvironmental subsets were shared across different patients. Clustering data showed significant intratumoural heterogeneity. The results also revealed a subgroup of cells bridging the metastatic group and primary group, implying the transition state of cancer during the metastatic process. In the present study, we obtained a more comprehensive picture of gastric cancer lymph node metastasis, and we discovered some GC lymph node metastasis marker genes (ERBB2, CLDN11 and CDK12), as well as potential gastric cancer evolution-driving genes (FOS and JUN), which provide a basis for the treatment of GC.


Assuntos
Metástase Linfática/genética , Neoplasias Gástricas/genética , Idoso , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática/patologia , Masculino , Pessoa de Meia-Idade , RNA-Seq , Análise de Célula Única , Neoplasias Gástricas/patologia
18.
Anticancer Agents Med Chem ; 21(16): 2192-2197, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-33397273

RESUMO

BACKGROUND: Neurokinin-1 receptor antagonists are playing a major advance in Chemotherapy-Induced Nausea and Vomiting (CINV) as powerful prophylactic agents. Therefore, it is significant to find the association between risk factors of patients and CINV so as to adjust the anti-emetic regimens. OBJECTIVE: To evaluate the role of Neurokinin-1 Receptor (NK-1R) antagonist in preventing chemotherapy-induced vomiting in the acute and delayed phases following the first cycle of treatment. METHODS: 145 adult patients with various cancers were recruited in Shanghai Changhai Hospital between September 2017 to November 2017, receiving dual or triple antiemetics. RESULTS: NK-1R antagonist combined with dexamethasone, 5-HT3R antagonist could effectively control delayed- vomiting in patients after Cycle 1 chemotherapy treatment (4.1% vs. 15.6%, P = 0.041<; 0.05). This study also showed that a history of motion sickness was a predictor of chemotherapy-induced vomiting (CINV) (P = 0.023 <; 0.05). In delayed phase a low consumption of alcohol and history of CIV for males were also significantly associated with CINV (P = 0.036 <; 0.05 and P = 0.002 <; 0.05 respectively). CONCLUSION: In this study, we found that triple anti-emetic regimen with NK-1R antagonist could effectively prevent the delayed-vomiting than dual agents. Moreover, some risk factors were observed to be associated with CINV in the delayed phase.


Assuntos
Dexametasona/farmacologia , Neoplasias/tratamento farmacológico , Antagonistas dos Receptores de Neurocinina-1/farmacologia , Vômito/tratamento farmacológico , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Feminino , Humanos , Masculino , Neoplasias/induzido quimicamente , Vômito/induzido quimicamente
19.
Oncol Lett ; 20(6): 276, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33029204

RESUMO

The prognosis of invasive pancreatic mucinous cystadenocarcinoma (MCC) is poor, and the molecular mechanism underlying its development remains unclear. The present study aimed to explore the potential role of autophagy in pancreatic MCC. The results demonstrated an increase in autophagy signaling in pancreatic MCC tissues and the MCC1 cell line compared with adjacent tissues and normal human pancreatic ductal epithelium (HPDE) cells. In addition, abnormal autophagy activation facilitated the migration and invasion of MCC1 cells. MicroRNA (miR)-224-5p expression levels were significantly higher in MCC1 cells compared with those in HPDE cells. Treatment with rapamycin further demonstrated that high levels of autophagy elevated miR-224-5p expression in MCC1 cells in a time-dependent manner. BCL2 was identified as a downstream target gene of miR-224-5p, which binds to the 3'-untranslated region of BCL2. In addition, the results of the present study demonstrated that BCL2 knockdown reversed the inhibition of autophagy mediated by the miR-224-5p inhibitor. To the best of our knowledge, this is the first study to evaluate the role of autophagy in pancreatic MCC. Thus, these results suggested that autophagy may be hyperactivated in pancreatic MCC. In addition, the present study identified a positive feedback loop between autophagy signaling and miR-224-5p, which may promote the aggressive migration and invasion of MCC1. These results may provide a new insight into the relationship between autophagy and tumor metastasis in pancreatic MCC.

20.
Cancer Cell Int ; 20: 339, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32760217

RESUMO

BACKGROUND: GATA binding protein 3 (GATA3) and miR-29b are related to colorectal cancer (CRC). The current study explored the regulatory relationship between GATA3 and miR-29b, and the mechanism of the two in the drug resistance of CRC cells to oxaliplatin. METHOD: Apoptosis of CRC cells induced by oxaliplatin at various doses was detected by flow cytometry. CRC cells were separately transfected with overexpression and knockdown of GATA3, miR-29b agomir and antagomir, and treated by oxaliplatin to detect the cell viability and apoptosis by performing Cell Couting Kit-8 (CCK-8) and flow cytometry. The expression levels of GATA3, caspase3 and cleaved caspase3 were determined by Western blot, and the expression of miR-29b was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Animal experiments were performed to examine the changes of transplanted tumors in nude mouse xenograft studies and observed by in vivo imaging. TUNEL staining was performed to detect tumor cell apoptosis. RESULT: Both GATA3 and miR-29b agomir inhibited the activity of the CRC cells, promoted apoptosis and Cleaved caspase3 expression, and reduced the resistance of the cells to chemotherapy drug oxaliplatin. Although GATA3 could up-regulate miR-29b expression, the tumor-suppressive effect of GATA3 was partially reversed by miR-29b antagomir. In vivo experiments showed that down-regulating the expression of GATA3 promoted the growth rate and volume of transplanted tumors, while overexpressing GATA3 had no significant effect on tumor growth. TUNEL staining results showed that knocking down or overexpression of GATA3 did not cause significant changes to apoptotic bodies of CRC cells, while oxaliplatin treatment increased the number of apoptotic bodies. CONCLUSION: GATA3 inhibits the cell viability of CRC cells, promotes apoptosis, and reduces oxaliplatin resistance of CRC cells through regulating miR-29b.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...