Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Int J Oncol ; 64(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38695252

ABSTRACT

Tumor­associated macrophages (TAMs) are essential components of the tumor microenvironment (TME) and display phenotypic heterogeneity and plasticity associated with the stimulation of bioactive molecules within the TME. TAMs predominantly exhibit tumor­promoting phenotypes involved in tumor progression, such as tumor angiogenesis, metastasis, immunosuppression and resistance to therapies. In addition, TAMs have the potential to regulate the cytotoxic elimination and phagocytosis of cancer cells and interact with other immune cells to engage in the innate and adaptive immune systems. In this context, targeting TAMs has been a popular area of research in cancer therapy, and a comprehensive understanding of the complex role of TAMs in tumor progression and exploration of macrophage­based therapeutic approaches are essential for future therapeutics against cancers. The present review provided a comprehensive and updated overview of the function of TAMs in tumor progression, summarized recent advances in TAM­targeting therapeutic strategies and discussed the obstacles and perspectives of TAM­targeting therapies for cancers.


Subject(s)
Disease Progression , Neoplasms , Tumor Microenvironment , Tumor-Associated Macrophages , Humans , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Neoplasms/drug therapy , Neovascularization, Pathologic/immunology , Animals , Molecular Targeted Therapy/methods
2.
World J Surg Oncol ; 22(1): 117, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38698475

ABSTRACT

BACKGROUND AND AIMS: The prevalence of metabolic dysfunction-associated fatty liver disease (MAFLD) in hepatocellular carcinoma (HCC) patients is increasing, yet its association with postoperative complications of HCC remains unclear. The aim of this study was to investigate the impact of MAFLD on complications after radical resection in HCC patients. METHODS: Patients with HCC who underwent radical resection were included. Patients were stratified into MAFLD group and non-MAFLD group. Clinical features and post-hepatectomy complications were compared between the two groups, and logistic regression analysis was used to determine independent risk factors associated with post-hepatectomy complications. RESULTS: Among the 936 eligible patients with HCC who underwent radical resection, concurrent MAFLD was diagnosed in 201 (21.5%) patients. Compared to the non-MAFLD group, the MAFLD group exhibited a higher incidence of complications, including infectious and major complications after radical resection in HCC patients. The logistic regression analysis found that MAFLD was an independent risk factor for complications, including infectious and major complications in HCC patients following radical resection (OR 1.565, 95%CI 1.109-2.343, P = 0.012; OR 2.092, 95%CI 1.386-3.156, P < 0.001; OR 1.859, 95% CI 1.106-3.124, P = 0.019; respectively). Subgroup analysis of HBV-related HCC patients yielded similar findings, and MAFLD patients with type 2 diabetes mellitus (T2DM) exhibited a higher incidence of postoperative complications compared to those without T2DM (all P < 0.05). CONCLUSIONS: Concurrent MAFLD was associated with an increased incidence of complications after radical resection in patients with HCC, especially MAFLD with T2DM.


Subject(s)
Carcinoma, Hepatocellular , Hepatectomy , Liver Neoplasms , Postoperative Complications , Humans , Carcinoma, Hepatocellular/surgery , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Male , Liver Neoplasms/surgery , Liver Neoplasms/pathology , Female , Postoperative Complications/etiology , Postoperative Complications/epidemiology , Middle Aged , Hepatectomy/adverse effects , Risk Factors , Follow-Up Studies , Prognosis , Retrospective Studies , Fatty Liver/etiology , Fatty Liver/epidemiology , Fatty Liver/complications , Fatty Liver/metabolism , Fatty Liver/pathology , Aged , Incidence
3.
Biomarkers ; 29(4): 211-221, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38629165

ABSTRACT

BACKGROUND: Increased lactate levels and metastasis in tumours are strongly associated with dismal outcomes. But prognostic value of lactate metabolism and transport-related lncRNAs in gastric adenocarcinoma (GA) patients remains unaddressed. METHODS: Gene expression data of GA were provided by The Cancer Genome Atlas. Lactate metabolism and transport-related gene data were accessed from GSEA. LncRNAs related to lactate metabolism and transport were identified by correlation analysis. A prognostic model was built by regression analysis. Validity of prognostic model was confirmed through survival analysis and receiver operating characteristic (ROC) curve. Immunity of each risk group was evaluated by immune correlation analysis .LncRNA-mRNA network was built by correlation analysis using Cytoscape software. RESULTS: A 12-gene prognostic model based on lactate metabolism and transport-related lncRNAs was built in GA. Median riskscore was utilized to classify GA samples into high- and low-risk groups. Survival analysis and ROC curves demonstrated validity of prognostic model. Most immune checkpoint molecules and TIDE scores were lower in the low-risk group. LINC01303 and LINC01545 may be the key prognostic factors in patients with GA. CONCLUSION: This study successfully built a prognostic model of lactate metabolism and transport-related lncRNAs in GA. The findings guide prognostic management of GA patients.


Subject(s)
Adenocarcinoma , Lactic Acid , RNA, Long Noncoding , Stomach Neoplasms , Humans , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Prognosis , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Lactic Acid/metabolism , Gene Expression Regulation, Neoplastic , ROC Curve , Biomarkers, Tumor/genetics , Male , Female , Survival Analysis
4.
Cancer Biol Ther ; 25(1): 2299288, 2024 12 31.
Article in English | MEDLINE | ID: mdl-38178596

ABSTRACT

Gastric cancer (GC) has been a major health burden all over the world but there are fewer promising chemotherapeutic drugs due to its multidrug resistance. It has been reported that WYC-209 suppresses the growth and metastasis of tumor-repopulating cells but the effect on GC was not explored. MTT, colony formation, and transwell assays were performed to examine the effects of WYC-209 on the proliferation, colony growth, and mobility of GC cells. Western blotting and qRT-PCR were used to detect the expression of proteins and mRNA. RNA-seq and enrichment analyses were conducted for the differentially expressed genes and enriched biological processes and pathways. The rescue experiments were carried out for further validation. Besides, we constructed xenograft model to confirm the effect of WYC-209 in vivo. The dual-luciferase reporter and Chromatin immunoprecipitation were implemented to confirm the underlying mechanism. WYC-209 exerted excellent anti-cancer effects both in vitro and in vivo. Based on RNA-seq and enrichment analyses, we found that Wnt family member 4 (WNT4) was significantly down-regulated. More importantly, WNT4 overexpression breached the inhibitory effect of WYC-209 on GC progression. Mechanically, WYC-209 significantly promoted the binding between retinoic acid receptor α (RARα) and WNT4 promoter. WYC-209 exerts anti-tumor effects in GC by down-regulating the expression of WNT4 via RARα.


Subject(s)
MicroRNAs , Stomach Neoplasms , Animals , Humans , Stomach Neoplasms/pathology , Cell Proliferation/genetics , Disease Models, Animal , Cell Line, Tumor , MicroRNAs/genetics , Gene Expression Regulation, Neoplastic , Cell Movement , Wnt4 Protein/genetics , Wnt4 Protein/metabolism
5.
Biochem Genet ; 62(1): 294-310, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37335371

ABSTRACT

To clarify the relationship between CD74 and atherosclerosis (AS) and the mechanisms in oxidized LDL (ox-LDL)-induced endothelial cell and macrophage injury. Datasets obtained from the Gene Expression Omnibus database are integrated. Differentially expressed genes (DEGs) were obtained using R software. Weighted gene co-expression network analysis (WGCNA) was performed to screen the target genes. The endothelial cell injury model and macrophage foaming model were established using ox-LDL, and CD74 expression was detected by Quantitative reverse transcription PCR (RT-qPCR) and Western blot (WB). Then, after silencing CD74, cell viability and ROS production were measured, and WB detected the expression of p-p38 MAPK and NF-κB. There were 268 DEGs associated with AS, of which CD74 was up-regulated. The turquoise module containing CD74 in WGCNA was positively associated with AS. Cell viability was significantly decreased in the endothelial cell injury and macrophage foaming models, while CD74, ROS production, NF-κB, and p-p38MAPK expression increased (P < 0.05). After silencing CD74, ROS production, NF-κB, and p-p38MAPK expression were decreased and cell viability was higher than the model group (P < 0.05). CD74 is up-regulated in endothelial cell injury and macrophage foaming models and is involved in AS progression via the NF-κB and MAPK signaling pathways.


Subject(s)
Atherosclerosis , NF-kappa B , Humans , Atherosclerosis/genetics , Endothelial Cells/metabolism , Lipoproteins, LDL/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism
6.
Cancer Rep (Hoboken) ; 6(10): e1888, 2023 10.
Article in English | MEDLINE | ID: mdl-37688511

ABSTRACT

BACKGROUND: Patients with combined hepatocellular-cholangiocarcinoma (cHCC-CCA) have limited treatment options and poor prognosis. Tumor-associated macrophages (TAMs) are the most abundant infiltrating immune cells in the tumor microenvironment and promote tumor stemness, proliferation, invasion and metastasis. Evidence suggested that transthyretin (TTR) influenced the prolifetation and invasion functions of different tumors and play an essential role in the tumor microenvironment. AIMS: To investigate the involvement of TTR in TAMs affecting the invasion of cHCC-CCA. METHODS AND RESULTS: Data sets obtained from the Gene Expression Omnibus database were integrated. Differentially expressed genes (DEGs) were obtained using R software, and modules associated with cHCC-CCA were screened by weighted gene co-expression network analysis (WGCNA). Human THP-1 cells were induced to differentiate into macrophages and then co-cultured with HCCC9810 cells and tumor necrosis factor-α (TNF-α) to simulate the inflammatory microenvironment of cHCC-CAA. In addition, small interfering RNA against TTR was transfected into HCCC9810 cells, and recombinant TTR and ERK and AKT-specific inhibitors were added to HCCC9810 cells, respectively; after that, the levels of NF-κB protein and phosphorylated ERK and AKT were measured. The invasive abilities of HCCC9810 cells were also tested. One hundred forty-five DEGs were associated with cHCC-CCA, of which TTR was up-regulated. Turquoise modules containing TTR in WGCNA were most significantly associated with cHCC-CCA. TTR was highly expressed in HCCC9810 compared to Huh-28. HCCC9810 showed enhanced invasive capacity after co-culture with TNF-α + macrophages (p < .05). After interfering with TTR, the invasive ability of HCCC9810 was diminished, accompanied by decreased expression of NF-κB, p-ERK1/2, and p-AKT (p < .05). After treating HCCC9810 with ERK and AKT-specific inhibitors, the invasive ability of HCCC9810 was diminished, accompanied by decreased expression of NF-κB and TTR (p < .05). CONCLUSION: TTR can promote the invasive ability of cHCC-CCA by regulating AKT/NF-κB and ERK pathways with the assistance of TAMs.


Subject(s)
Bile Duct Neoplasms , Carcinoma, Hepatocellular , Cholangiocarcinoma , Liver Neoplasms , Humans , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Carcinoma, Hepatocellular/genetics , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Liver Neoplasms/genetics , NF-kappa B , Prealbumin/genetics , Proto-Oncogene Proteins c-akt/metabolism , Tumor Microenvironment , Tumor Necrosis Factor-alpha , Tumor-Associated Macrophages/metabolism
7.
Cancer Lett ; 574: 216391, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37714257

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly life-threatening tumour with a low early-detection rate, rapid progression and a tendency to develop resistance to chemotherapy. Therefore, understanding the regulatory mechanisms underlying the initiation, development and metastasis of pancreatic cancer is necessary for enhancing therapeutic effectiveness. In this review, we summarised single-gene mutations (including KRAS, CDKN2A, TP53, SMAD4 and some other less prevalent mutations), epigenetic changes (including DNA methylation, histone modifications and RNA interference) and large chromosome alterations (such as copy number variations, chromosome rearrangements and chromothripsis) associated with PDAC. In addition, we discussed variations in signalling pathways that act as intermediate oncogenic factors in PDAC, including PI3K/AKT, MAPK/ERK, Hippo and TGF-ß signalling pathways. The focus of this review was to investigate alterations in the microenvironment of PDAC, particularly the role of immunosuppressive cells, cancer-associated fibroblasts, lymphocytes, other para-cancerous cells and tumour extracellular matrix in tumour progression. Peripheral axons innervating the pancreas have been reported to play a crucial role in the development of cancer. In addition, tumour cells can influence the behaviour of neighbouring non-tumour cells by secreting certain factors, both locally and at a distance. In this review, we elucidated the alterations in intracellular molecules and the extracellular environment that occur during the progression of PDAC. Altogether, this review may enhance the understanding of the biological characteristics of PDAC and guide the development of more precise treatment strategies.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , DNA Copy Number Variations , Phosphatidylinositol 3-Kinases/genetics , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Tumor Microenvironment , Pancreatic Neoplasms
8.
Brachytherapy ; 22(6): 858-871, 2023.
Article in English | MEDLINE | ID: mdl-37574351

ABSTRACT

BACKGROUND: Therapy for hepatocellular carcinoma (HCC) patients with portal vein tumor thrombosis (PVTT) is still controversial. This study was performed to evaluate the efficacy and safety of the combination therapy comprising transarterial chemoembolization (TACE), lenvatinib (L), programmed death-1 inhibitor (P), and iodine-125 seed (I125) brachytherapy relative to TACE in combination with lenvatinib plus programmed death-1 inhibitor therapy and TACE plus lenvatinib therapy. METHODS: The data of HCC patients with PVTT from July 2017 to August 2022 were assessed in this single-center retrospective study. Primary study outcomes were progression-free survival (PFS) and overall survival (OS), while the secondary outcomes were disease control rate (DCR), objective response rate (ORR), and treatment-related adverse events. RESULTS: We enrolled 150 patients totally, including 50 patients treated with TACE plus lenvatinib therapy (TACE+L group), 45 patients treated with TACE in combination with lenvatinib plus programmed death-1 inhibitor therapy (TACE+L+P group), and 55 patients treated with the combination therapy of TACE along with I125 brachytherapy, lenvatinib, and programmed death-1 inhibitor therapy (TACE+L+P+I125 group). The median OS in the TACE+L+P+I125 group (21.0; 95% confidence interval [CI]: 18.4∼23.5 months) was significantly longer than that in the TACE+L group (10; 95% CI: 7.8∼12.1months) (p = 0.006), while it was insignificantly longer than that in the TACE+L+P group (14.0; 95% CI: 10.7∼17.2months) (p = 0.058). The median PFS in the TACE+L+P+I125 group (13.0; 95% CI: 10.2∼15.7 months) was significantly longer than that in the TACE+L group (5.0; 95% CI: 4.2∼5.7 months) (p = 0.014) and the TACE+L+P group (9.0; 95% CI: 6.7∼11.2 months) (p = 0.048). Statistically significant differences between groups were found in DCR (p = 0.015). There were no significant between-group differences in treatment-related adverse events (p > 0.05). CONCLUSIONS: A combination therapy of TACE, lenvatinib, programmed death-1 inhibitor, and I125 seed brachytherapy significantly improve OS, PFS, and DCR and show better survival prognosis for HCC patients accompanied by PVTT.


Subject(s)
Brachytherapy , Carcinoma, Hepatocellular , Chemoembolization, Therapeutic , Iodine Radioisotopes , Liver Neoplasms , Phenylurea Compounds , Quinolines , Thrombosis , Humans , Carcinoma, Hepatocellular/radiotherapy , Liver Neoplasms/radiotherapy , Brachytherapy/methods , Portal Vein , Retrospective Studies , Seeds
9.
Front Oncol ; 13: 1178428, 2023.
Article in English | MEDLINE | ID: mdl-37207144

ABSTRACT

Background: The subsequent therapy for hepatocellular carcinoma (HCC) patients with refractory to transarterial chemoembolization (TACE) is still controversial. This study was performed to evaluate the efficacy and safety of combination therapy comprising hepatic artery infusion chemotherapy (HAIC), lenvatinib, and programmed death-1 inhibitors relative to HAIC combined with lenvatinib. Methods: In this single-center retrospective study, we analyzed data from HCC patients with refractory to TACE from June 2017 to July 2022. Primary study outcomes were overall survival (OS) and progression-free survival (PFS), while the secondary outcomes were the objective response rate (ORR), disease control rate (DCR), and treatment-related adverse events. Results: We enrolled 149 patients finally, including 75 patients who received HAIC combined with lenvatinib plus PD-1 inhibitors therapy (HAIC+L+P group) and 74 patients who received HAIC combined with lenvatinib therapy (HAIC+L group). The median OS in the HAIC+L+P group (16.0; 95% CI: 13.6~18.3 months) was significantly higher compared to the HAIC+L group (9.0; 95% CI: 6.5~11.4 months) (p = 0.002), while the median PFS in the HAIC+L+P group (11.0; 95% CI: 8.6~13.3 months) was significantly higher compared to the HAIC+L group (6.0; 95% CI: 5.0~6.9 months) (p < 0.001). Significant between-group differences in DCR (p = 0.027) were found. Additionally, 48 pairs of patients were matched after propensity matching analysis. The survival prognosis between two groups before propensity matching is similar to that after propensity matching. Moreover, the percentage of patients with hypertension in the HAIC+L+P group was significantly higher compared to the HAIC+L group (28.00% vs. 13.51%; p = 0.029). Conclusions: A combination therapy of HAIC, lenvatinib, and programmed death-1 inhibitors significantly improved oncologic response and prolonged survival duration, showing a better survival prognosis for HCC patients with refractory toTACE.

10.
Medicine (Baltimore) ; 102(9): e33062, 2023 Mar 03.
Article in English | MEDLINE | ID: mdl-36862923

ABSTRACT

Metabolic dysfunction-associated fatty liver disease (MAFLD) is a term that was proposed in 2020 by a group of international experts. However, the impact of MAFLD on complications after hepatectomy in patients with hepatocellular carcinoma is not clear. The aim of this study is to explore the influence of MAFLD on the complications after hepatectomy in patients with hepatitis B virus-related hepatocellular carcinoma (HBV-HCC). Patients with HBV-HCC who underwent hepatectomy between January 2019 and December 2021 were consecutively enrolled. The predictors of complications after hepatectomy in HBV-HCC patients were retrospectively analyzed. Among the 514 eligible HBV-HCC patients, 117 (22.8%) were diagnosed with concurrent MAFLD. Post hepatectomy complications occurred in 101 patients (19.6%), including 75 patients (14.6%) with infectious complications and 40 patients (7.8%) with major complications. Univariate analysis showed that MAFLD was not the risk factor for complications after hepatectomy in patients with HBV-HCC (P > .05). However, univariate and multivariate analysis revealed that lean-MAFLD was an independent risk factor for post hepatectomy complications in patients with HBV-HCC (odds ratio 2.245; 95% confidence interval 1.243-5.362, P = .028). Similar results were found in the analysis of predictors for infectious and major complications after hepatectomy in patients with HBV-HCC. MAFLD commonly coexists with HBV-HCC and is not directly associated with complications after hepatectomy, but lean-MAFLD is an independent risk factor for post hepatectomy complications in patients with HBV-HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Carcinoma, Hepatocellular/surgery , Hepatitis B virus , Hepatectomy/adverse effects , Retrospective Studies , Liver Neoplasms/surgery
11.
Hepatobiliary Pancreat Dis Int ; 22(4): 366-372, 2023 Aug.
Article in English | MEDLINE | ID: mdl-35466065

ABSTRACT

BACKGROUND: Metabolic dysfunction-associated fatty liver disease (MAFLD) is recently proposed an entity by a group of international experts. However, the impact of MAFLD on the prognosis of patients with hepatocellular carcinoma (HCC) is not clear. The aim of this study was to explore the influence of MAFLD for the prognosis of HCC after radical resection. METHODS: HCC patients who received radical resection were enrolled. The recurrence-free survival (RFS) and overall survival (OS) were compared between MAFLD and non-MAFLD. RESULTS: A total of 576 HCC patients were included, and among them 114 (19.8%) met the diagnostic criteria of MAFLD. The median RFS was 34.0 months in the MAFLD group and 19.0 months in the non-MAFLD group. The 1-, 3-, and 5-year RFS rates were 64.9%, 49.1% and 36.1% in the MAFLD group, which were higher than those of the non-MAFLD group (59.4%, 35.3% and 26.5%, respectively, P = 0.01). The mean OS was 57.0 months in the MAFLD group and 52.2 months in the non-MAFLD group. There was no statistical difference in OS rate between the MAFLD group and non-MAFLD group. Similar results were found in HBV-related HCC patients in the subgroup analysis. Univariate analysis revealed that MAFLD was a protective factor for RFS in HCC patients after radical resection (P < 0.05), and there was no association between MAFLD and OS rate (P > 0.05). Multivariate analysis demonstrated that MAFLD was not an independent protective factor for HCC patients with radical resection. CONCLUSIONS: MAFLD improves RFS rate in HCC patients with radical resection, but is not an independent protective factor and not associated with OS rate.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Retrospective Studies , Prognosis , Hepatectomy/adverse effects
12.
Front Immunol ; 14: 1286771, 2023.
Article in English | MEDLINE | ID: mdl-38288113

ABSTRACT

Objectives: To evaluate the efficacy and safety of biliary stenting implantation with iodine-125 seed strand (SI) followed by hepatic artery infusion chemotherapy (HAIC) plus lenvatinib (Len) with programmed death-1 (PD-1) inhibitor for patients diagnosed with extrahepatic cholangiocarcinoma (ECC) and malignant obstructive jaundice (MOJ). Methods: In this single-center retrospective study, the data of ECC patients with MOJ from March 2015 to January 2023 was assessed. Using probability score matching (PSM), the selection bias of patients was reduced. Primary study outcomes included overall survival (OS) and progression-free survival (PFS). The OS and PFS were performed using the Kaplan-Meier method and evaluated with the log-rank test. Results: A total of 104 patients were enrolled finally, including 52 patients treated with interventional therapy (SI+HAIC) plus Len with PD-1 inhibitor (SI+HAIC+Len+P group) and 52 patients treated with interventional therapy (SI+HAIC) plus lenvatinib (SI+HAIC+Len group). 26 pairs of patients were matched after PSM analysis. After PSM analysis, the median OS and PFS in the SI+HAIC+Len+P group were significantly longer compared to those in the SI+HAIC+Len group (OS:16.6 vs. 12.3 months, P = 0.001; PFS:12.6 vs 8.5 months, P = 0.004). The DCR was significantly different between groups (P = 0.039), while ORR not (P = 0.548). The addition of PD-1 inhibitor was generally well tolerated without treatment-associated mortality. Conclusion: Interventional therapy (SI+HAIC) plus Len with PD-1 inhibitor was effective for ECC patients accompanied by MOJ with a manageable safety profile.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Iodine Radioisotopes , Jaundice, Obstructive , Phenylurea Compounds , Quinolines , Humans , Immune Checkpoint Inhibitors , Hepatic Artery , Retrospective Studies , Cholangiocarcinoma/complications , Cholangiocarcinoma/therapy , Bile Duct Neoplasms/complications , Bile Duct Neoplasms/therapy , Bile Ducts, Intrahepatic
13.
Front Pharmacol ; 13: 919819, 2022.
Article in English | MEDLINE | ID: mdl-36046825

ABSTRACT

Macropinocytosis, a unique endocytosis pathway characterized by nonspecific internalization, has a vital role in the uptake of extracellular substances and antigen presentation. It is known to have dual effects on cancer cells, depending on cancer type and certain microenvironmental conditions. It helps cancer cells survive in nutrient-deficient environments, enhances resistance to anticancer drugs, and promotes invasion and metastasis. Conversely, overexpression of the RAS gene alongside drug treatment can lead to methuosis, a novel mode of cell death. The survival and proliferation of cancer cells is closely related to macropinocytosis in the tumor microenvironment (TME), but identifying how these cells interface with the TME is crucial for creating drugs that can limit cancer progression and metastasis. Substantial progress has been made in recent years on designing anticancer therapies that utilize the effects of macropinocytosis. Both the induction and inhibition of macropinocytosis are useful strategies for combating cancer cells. This article systematically reviews the general mechanisms of macropinocytosis, its specific functions in tumor cells, its occurrence in nontumor cells in the TME, and its application in tumor therapies. The aim is to elucidate the role and therapeutic potential of macropinocytosis in cancer treatment.

14.
Front Oncol ; 12: 916082, 2022.
Article in English | MEDLINE | ID: mdl-36033459

ABSTRACT

Ferroptosis is an iron-dependent cell death process characterized by excessive accumulation of reactive oxygen species and lipid peroxidation. The elucidation of ferroptosis pathways may lead to novel cancer therapies. Current evidence suggests that the mechanism of ferroptosis can be summarized as oxidative stress and antioxidant defense mechanisms. During this process, ferrous ions play a crucial role in cellular oxidation, plasma membrane damage, reactive oxygen species removal imbalance and lipid peroxidation. Although, disregulation of intracellular cations (Fe2+, Ca2+, Zn2+, etc.) and anions (Cl-, etc.) have been widely reported to be involved in ferroptosis, their specific regulatory mechanisms have not been established. To further understand the crosstalk effect between ferrous and other ions in ferroptosis, we reviewed the ferroptosis process from the perspective of ions metabolism. In addition, the role of ferrous and other ions in tumor therapy is briefly summarized.

15.
Front Immunol ; 13: 887048, 2022.
Article in English | MEDLINE | ID: mdl-35784334

ABSTRACT

Background: Colorectal cancer (CRC) is one of the most common malignancies and its incidence and mortality are increasing yearly. 5-Fluorouracil (5-FU) has long been used as a standard first-line treatment for CRC patients. Although 5-FU-based chemotherapy is effective for advanced CRC, the consequent resistance remains a key problem and causes the poor prognosis of CRC patients. Thus, there is an urgent need to identify new biomarkers to predict the response to 5-FU-based chemotherapy. Methods: CRC samples were retrieved from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA). The immune-related genes were retrieved from the ImmPort database. Single-cell sequencing results from colorectal cancer were obtained by the ArrayExpress database. 5-FU resistance-related genes were filtered and validated by R packages. ESTIMATE algorithms were used to assess the tumor microenvironment (TME). KEGG and GO analysis were performed to explore the biological signaling pathway for resistant-response patients and sensitive-response patients in the tumor microenvironment. pRRophetic algorithms were used to predict 5-FU sensitivity. GSEA and GSVA analysis was performed to excavate the biological signaling pathway of the RBP7 gene. Results: Nine immune-related genes were identified to be associated with 5-FU resistance and poor disease-free survival (DFS) of CRC patients and the signature of these genes was developed in a DFS-prognostic model. Four immune-related genes were determined to be associated with 5-FU resistance and overall survival (OS) of CRC patients. The signature of these genes was developed an OS-prognostic model. ESTIMATE scores showed a significant difference between 5-FU resistant and 5-FU sensitive CRC patients. Resistant-response patients and sensitive-response patients to 5-FU based chemotherapy showed different GO and KEGG enrichment on the tumor microenvironment. RBP7, as a tumor immune microenvironment (TIME) related gene, was found to have the potential of predicting chemotherapy resistance and poor prognosis of CRC patients. GSEA analysis showed multiple signaling differences between the high and low expression of RBP7 in CRC patients. Hypoxia and TNFα signaling via NFκB gene sets were significantly different between chemotherapy resistant (RBP7High) and chemotherapy sensitive (RBP7Low) patients. Single-cell RNA-seq suggested RBP7 was centrally distributed in endothelial stalk cells, endothelial tip cells, and myeloid cells. Conclusions: Immune-related genes will hopefully be potential prognostic biomarkers to predict chemotherapy resistance for CRC. RBP7 may function as a tumor microenvironment regulator to induce 5-FU resistance, thereby affecting the prognosis of CRC patients.


Subject(s)
Colorectal Neoplasms , Fluorouracil , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Tumor Microenvironment/genetics
16.
Transl Cancer Res ; 11(1): 99-112, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35261888

ABSTRACT

Background: The majority of patients with hepatocellular carcinoma (HCC) are diagnosed in an advanced stage. Although sorafenib is recommended as the standard treatment for advanced HCC, its efficacy is limited. In some studies, hepatic arterial infusion chemotherapy has demonstrated a significant therapeutic benefit for advanced HCC compared with sorafenib. We systematically evaluated and compared the efficacy and safety of hepatic arterial infusion chemotherapy and sorafenib for advanced HCC. Methods: A systematic search of PubMed, Embase, Web of Science and the Cochrane Library up to December 31, 2020 was conducted. Study outcomes included overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and adverse effects. The hazard ratio and odds ratio with 95% confidence intervals (CI) were used to measure the pooled effect. Results: Eighteen retrospective or prospective cohort studies and one prospective controlled study were included, with 1,339 patients treated with hepatic arterial infusion chemotherapy (HAIC) and 1,060 patients treated with sorafenib. We found that hepatic arterial infusion chemotherapy was superior to sorafenib in terms of OS [hazard ratio (HR): 0.66, 95% CI: 0.46-0.95, P=0.027], PFS (HR: 0.55, 95% CI: 0.44-0.69, P<0.001), ORR [assessed using Response Evaluation Criteria in Solid Tumors (RECIST): OR: 9.02, 95% CI: 6.01-13.53, P<0.001; assessed using modified RECIST: odds ratio (OR): 3.71, 95% CI: 1.92-7.16, P<0.001], and DCR (assessed using RECIST: OR: 2.31, 95% CI: 1.40-3.83, P=0.001; assessed by modified RECIST: OR: 2.28, 95% CI: 1.22-4.28, P=0.01). Dermatological adverse effects and hypertension were significantly higher in the sorafenib group for all grades of adverse effects. However, regarding severe adverse effects, hepatic arterial infusion chemotherapy was associated with more frequent leukocytopenia and thrombocytopenia. Conclusions: Hepatic arterial infusion chemotherapy demonstrated favorable efficacy and safety for advanced HCC compared with sorafenib and should be recommended for suitable patients with advanced HCC.

17.
IEEE Trans Image Process ; 31: 2122-2135, 2022.
Article in English | MEDLINE | ID: mdl-35196236

ABSTRACT

Multi-source domain adaptation (MDA) aims to transfer knowledge from multiple source domains to an unlabeled target domain. MDA is a challenging task due to the severe domain shift, which not only exists between target and source but also exists among diverse sources. Prior studies on MDA either estimate a mixed distribution of source domains or combine multiple single-source models, but few of them delve into the relevant information among diverse source domains. For this reason, we propose a novel MDA approach, termed Pseudo Target for MDA (PTMDA). Specifically, PTMDA maps each group of source and target domains into a group-specific subspace using adversarial learning with a metric constraint, and constructs a series of pseudo target domains correspondingly. Then we align the remainder source domains with the pseudo target domain in the subspace efficiently, which allows to exploit additional structured source information through the training on pseudo target domain and improves the performance on the real target domain. Besides, to improve the transferability of deep neural networks (DNNs), we replace the traditional batch normalization layer with an effective matching normalization layer, which enforces alignments in latent layers of DNNs and thus gains further promotion. We give theoretical analysis showing that PTMDA as a whole can reduce the target error bound and leads to a better approximation of the target risk in MDA settings. Extensive experiments demonstrate PTMDA's effectiveness on MDA tasks, as it outperforms state-of-the-art methods in most experimental settings.


Subject(s)
Neural Networks, Computer
18.
IEEE Trans Cybern ; 52(8): 8352-8365, 2022 Aug.
Article in English | MEDLINE | ID: mdl-33544687

ABSTRACT

For a broad range of applications, hyperspectral image (HSI) classification is a hot topic in remote sensing, and convolutional neural network (CNN)-based methods are drawing increasing attention. However, to train millions of parameters in CNN requires a large number of labeled training samples, which are difficult to collect. A conventional Gabor filter can effectively extract spatial information with different scales and orientations without training, but it may be missing some important discriminative information. In this article, we propose the Gabor ensemble filter (GEF), a new convolutional filter to extract deep features for HSI with fewer trainable parameters. GEF filters each input channel by some fixed Gabor filters and learnable filters simultaneously, then reduces the dimensions by some learnable 1×1 filters to generate the output channels. The fixed Gabor filters can extract common features with different scales and orientations, while the learnable filters can learn some complementary features that Gabor filters cannot extract. Based on GEF, we design a network architecture for HSI classification, which extracts deep features and can learn from limited training samples. In order to simultaneously learn more discriminative features and an end-to-end system, we propose to introduce the local discriminant structure for cross-entropy loss by combining the triplet hard loss. Results of experiments on three HSI datasets show that the proposed method has significantly higher classification accuracy than other state-of-the-art methods. Moreover, the proposed method is speedy for both training and testing.


Subject(s)
Algorithms , Neural Networks, Computer
19.
Cancer Cell Int ; 21(1): 538, 2021 Oct 17.
Article in English | MEDLINE | ID: mdl-34657635

ABSTRACT

BACKGROUND: Increased tryptophan (Trp) metabolism by indoleamine 2,3-dioxygenase (IDO)/tryptophan 2,3-dioxygenase (TDO) represents one of the most studied pathways for immunosuppression in tumor tissues. However, the pro-tumor effects induced by Trp metabolism remain controversial. METHODS: The paraffin sections of tumor tissues were obtained from patients with liver cancer and examined by immunohistochemical staining to investigate the role of Trp metabolic enzymes. To further confirm the pro-tumor effects induced by TDO2, we established TDO2 overexpression SMC-7721 and HepG2 liver cancer cell lines, and western blotting, cell proliferation, and colony formation were evaluated. Meanwhile, liver cancer subcutaneous mice models were established, and the tumorigenic rates of SMC-7721 cells, tumor volume and survival of bearing mice were calculated. In addition, the survival data of liver cancer patients from The Cancer Genome Atlas (TCGA) database were downloaded to analyze the effect of TDO2 expression on the survival of patients with liver cancer. RESULTS: Here, we showed that constitutive TDO2 expression gave rise to liver cancer through upregulation of Trp metabolism. And the TDO2 expression was positively correlated with the poor prognosis in liver cancer patients. TDO2 expression in tumor cells accounted for the release of kynurenine (Kyn), which activated aryl hydrocarbon receptor (AhR) to promote liver cancer cells proliferation. Mechanistically, we found that AhR expression contributed to the secretion of Interleukin-6 (IL-6), thereby promoting tumor cells proliferation through the STAT3 and NF-kB/TIM4 signals. Interrupt of AhR signals by PDM2 revealed improved outcomes in subcutaneous tumor-bearing mice. CONCLUSIONS: Together, our study showed that the TDO2/Kyn/AhR/IL-6 signaling pathway was a novel mechanism underlying the malignancy of liver cancer, and suggested that AhR signals might be a valuable therapeutic target for tumor therapy.

20.
Front Pharmacol ; 12: 691769, 2021.
Article in English | MEDLINE | ID: mdl-34335258

ABSTRACT

Background: Hepatocellular carcinoma (HCC) is a lethal malignancy lacking effective treatment. The Cyclin-dependent kinases 4/6 (CDK4/6) and PI3K/AKT signal pathways play pivotal roles in carcinogenesis and are promising therapeutic targets for HCC. Here we identified a new CDK4/6 and PI3K/AKT multi-kinase inhibitor for the treatment of HCC. Methods: Using a repurposing and ensemble docking methodology, we screened a library of worldwide approved drugs to identify candidate CDK4/6 inhibitors. By MTT, apoptosis, and flow cytometry analysis, we investigated the effects of candidate drug in reducing cell-viability,inducing apoptosis, and causing cell-cycle arrest. The drug combination and thermal proteomic profiling (TPP) method were used to investigate whether the candidate drug produced antagonistic effect. The in vivo anti-cancer effect was performed in BALB/C nude mice subcutaneously xenografted with Huh7 cells. Results: We demonstrated for the first time that the anti-plasmodium drug aminoquinol is a new CDK4/6 and PI3K/AKT inhibitor. Aminoquinol significantly decreased cell viability, induced apoptosis, increased the percentage of cells in G1 phase. Drug combination screening indicated that aminoquinol could produce antagonistic effect with the PI3K inhibitor LY294002. TPP analysis confirmed that aminoquinol significantly stabilized CDK4, CDK6, PI3K and AKT proteins. Finally, in vivo study in Huh7 cells xenografted nude mice demonstrated that aminoquinol exhibited strong anti-tumor activity, comparable to that of the leading cancer drug 5-fluorouracil with the combination treatment showed the highest therapeutic effect. Conclusion: The present study indicates for the first time the discovery of a new CDK4/6 and PI3K/AKT multi-kinase inhibitor aminoquinol. It could be used alone or as a combination therapeutic strategy for the treatment of HCC.

SELECTION OF CITATIONS
SEARCH DETAIL
...