Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
Nat Commun ; 15(1): 1754, 2024 Feb 26.
Article En | MEDLINE | ID: mdl-38409200

The response to programmed death-1 (PD-1) blockade varies in hepatocellular carcinoma (HCC). We utilize a panel of 16 serum factors to show that a circulating level of serum amyloid A (SAA) > 20.0 mg/L has the highest accuracy in predicting anti-PD-1 resistance in HCC. Further experiments show a correlation between peritumoral SAA expression and circulating SAA levels in patients with progressive disease after PD-1 inhibition. In vitro experiments demonstrate that SAA induces neutrophils to express PD-L1 through glycolytic activation via an LDHA/STAT3 pathway and to release oncostatin M, thereby attenuating cytotoxic T cell function. In vivo, genetic or pharmacological inhibition of STAT3 or SAA eliminates neutrophil-mediated immunosuppression and enhances antitumor efficacy of anti-PD-1 treatment. This study indicates that SAA may be a critical inflammatory cytokine implicated in anti-PD-1 resistance in HCC. Targeting SAA-induced PD-L1+ neutrophils through STAT3 or SAA inhibition may present a potential approach for overcoming anti-PD1 resistance.


Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , B7-H1 Antigen/metabolism , Neutrophils/metabolism , Serum Amyloid A Protein/metabolism , Programmed Cell Death 1 Receptor , Glycolysis
2.
JHEP Rep ; 6(1): 100939, 2024 Jan.
Article En | MEDLINE | ID: mdl-38074509

Background & Aims: The mechanism underlying resistance to immunotherapy involves engagement of immune checkpoint pathways. The transcriptional and epigenetic processes of checkpoint molecules, however, have not been well investigated. We thus studied whether the transcription factor myeloid zinc finger 1 (MZF1) may promote resistance to immunotherapy in hepatocellular carcinoma (HCC). Methods: Single-cell RNA-sequencing was performed to study the correlation between MZF1 and tumour microenvironment features in six patients with HCC. Combined immunohistochemistry and multi-immunofluorescence analyses were performed for verification. Ectopic expression of MZF1 was used in both orthotopic and genetically engineered hydrodynamic mouse HCC models for in vivo experiments. Proteome analysis, including protein degradation assays, ubiquitination assays, and co-immunoprecipitation assays, revealed the function of MZF1 in immune checkpoint pathways. Results: Single-cell RNA-sequencing suggested an immunosuppressive environment and a strong correlation with the immune checkpoint programmed death ligand 1 (PD-L1) in MZF1-overexpressing tumours. Analyses of 163 HCC samples demonstrated that MZF1 expression in HCC cells is associated with decreased T-cell infiltration. In vivo experiments showed that ectopic MZF1 expression in HCC cells impairs T-cell recruitment, resulting in resistance to immune checkpoint blockade. Mechanistically, MZF1 accelerated PD-L1 ubiquitination by binding to the cyclin-dependent kinase 4 (CDK4) activation site, while a direct bond between CDK4 and MZF1 led to increased MZF1 expression. Conclusions: MZF1 promotes PD-L1 ubiquitination via CDK4 and possibly MZF1. Inhibition of CDK4 can therefore restore PD-L1 expression and may be a potential strategy for combination with anti-PD-L1 antibodies. Impact and implications: Resistance to immune checkpoint blockade with anti-programmed death ligand 1 (PD-L1) antibody therapy is attributed to oncogenic alterations of tumour cells, however, effective countermeasures are yet to be established. Here, we report that the transcription factor myeloid zinc finger 1 (MZF1) can bind to the cyclin-dependent kinase 4 (CDK4) activation site and accelerate PD-L1 ubiquitination. A CDK4 inhibitor therefore enhances anti-PD-L1 antibody efficacy by blocking MZF1 signalling. This indicates a potential benefit of combining CDK4 inhibitors and anti-PD-L1 antibodies for the treatment of advanced HCC.

3.
Sci Adv ; 9(18): eade5111, 2023 05 05.
Article En | MEDLINE | ID: mdl-37146141

Hypoxia-induced adenosine creates an immunosuppressive tumor microenvironment (TME) and dampens the efficacy of immune checkpoint inhibitors (ICIs). We found that hypoxia-inducible factor 1 (HIF-1) orchestrates adenosine efflux through two steps in hepatocellular carcinoma (HCC). First, HIF-1 activates transcriptional repressor MXI1, which inhibits adenosine kinase (ADK), resulting in the failure of adenosine phosphorylation to adenosine monophosphate. This leads to adenosine accumulation in hypoxic cancer cells. Second, HIF-1 transcriptionally activates equilibrative nucleoside transporter 4, pumping adenosine into the interstitial space of HCC, elevating extracellular adenosine levels. Multiple in vitro assays demonstrated the immunosuppressive role of adenosine on T cells and myeloid cells. Knockout of ADK in vivo skewed intratumoral immune cells to protumorigenic and promoted tumor progression. Therapeutically, combination treatment of adenosine receptor antagonists and anti-PD-1 prolonged survival of HCC-bearing mice. We illustrated the dual role of hypoxia in establishing an adenosine-mediated immunosuppressive TME and offered a potential therapeutic approach that synergizes with ICIs in HCC.


Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Mice, Knockout , Hypoxia/metabolism , Adenosine/metabolism , Cell Line, Tumor , Tumor Microenvironment
4.
Wei Sheng Yan Jiu ; 52(2): 253-258, 2023 Mar.
Article Zh | MEDLINE | ID: mdl-37062688

OBJECTIVE: To study the expression of heat shock protein 27(HSP27), BAX and BCL-2 apoptosis in silicosis rat model, and to explore the correlation between HSP27 and BAX and BCL-2 apoptosis. METHODS: Silicosis model was established by the oropharyngeal and endotracheal intubation. Forty SPF healthy adult Wistar male rats were randomly divided into 4 groups, with 10 rats in each group. Silicosis group for 6 weeks(feeding for 6 weeks), silicosis group for 8 weeks(feeding for 8 weeks): oropharyngeal and tracheal perfusion of 50 mg/mL SiO_2 suspension 1.0 mL/mouse; Model control group for 6 weeks and model control group for 8 weeks: 1.0 mL saline was infused into the oropharynx and trachea. Immunohistochemical staining was used to detect the expression of HSP27, BAX and BCL-2 in the right lower lung of silicosis model group at 6 and 8 weeks and model control group at 6 and 8 weeks. Western blot was used to detect the protein expression of HSP27, BAX and BCL-2 in the left lower lobe lung tissue of silicosis model group at 6 and 8 weeks and model control group at 6 and 8 weeks, respectively. Immunofluorescence staining was used to detect the colocalization of HSP27 with pro-apoptotic factor BAX and HSP27 with anti-apoptotic factor BCL-2. RESULTS: Compared with the model control group at 6 weeks and 8 weeks, the expression of HSP27 and pro-apoptotic factor BAX in fibrotic region increased, and the expression of anti-apoptotic factor BCL-2 decreased in silicosis model group at 6 weeks and 8 weeks(P<0.05). Immunofluorescence staining showed that there was colocalization of HSP27 and pro-apoptotic factor BAX in the fibrotic region. Correlation analysis showed that the correlation coefficient between HSP27 and pro-apoptotic factor BAX was r=0.94, indicating a positive correlation between them, while the correlation coefficient between HSP27 and anti-apoptotic factor BCL-2 was r=-0.81, indicating a negative correlation between them. CONCLUSION: High expression of HSP27 and pro-apoptotic factor BAX and low expression of anti-apoptotic factor BCL-2 exist in silicosis rats, and their expression is correlated.


HSP27 Heat-Shock Proteins , Silicosis , Rats , Male , Animals , Mice , Rats, Wistar , HSP27 Heat-Shock Proteins/genetics , bcl-2-Associated X Protein/metabolism , Proto-Oncogene Proteins c-bcl-2 , Apoptosis , Fibrosis
5.
J Clin Oncol ; 40(5): 468-480, 2022 02 10.
Article En | MEDLINE | ID: mdl-34905388

PURPOSE: Interventional hepatic arterial infusion chemotherapy of infusional fluorouracil, leucovorin, and oxaliplatin (HAIC-FO) displayed an encouraging safety profile and antitumor activity in a previous phase II trial and a propensity-score-matching study involving patients with locally advanced hepatocellular carcinoma (HCC). METHODS: In this open-label, phase III trial, patients with advanced HCC, previously untreated with systemic therapy, were randomly assigned in a 1:1 ratio to receive HAIC-FO or sorafenib. The primary end point was overall survival (OS) in the intention-to-treat population. An exploratory model for predicting the efficacy of HAIC-FO on the basis of genomic sequencing was developed. RESULTS: Between May 2017 and May 2020, 262 patients were randomly assigned. The median tumor size was 11.2 cm (interquartile range, 8.5-13.7 cm). Macrovascular invasion was present in 65.6%, and the percentage of patients with > 50% tumor volume involvement of the liver and/or Vp-4 portal vein tumor thrombosis was 49.2%. At data cutoff (October 31, 2020), median OS was 13.9 months for HAIC-FO and 8.2 for sorafenib (hazard ratio [HR] 0.408; 95% CI, 0.301 to 0.552; P < .001). Tumor downstaging occurred in 16 (12.3% of 130) patients receiving HAIC-FO, including 15 receiving curative surgery or ablation, and finally achieving a median OS of 20.8 months, with a 1-year OS rate of 93.8%. In high-risk subpopulations, OS was significantly longer with HAIC-FO than with sorafenib (10.8 months v 5.7 months; HR 0.343; 95% CI, 0.219 to 0.538; P < .001). A newly developed 15-mutant-gene prediction model identified 83% of patients with response to HAIC-FO. HAIC-FO responders had longer OS than HAIC-FO nonresponders (19.3 months v 10.6 months; HR 0.323; 95% CI, 0.186 to 0.560; P = .002). CONCLUSION: HAIC-FO achieved better survival outcomes than sorafenib in advanced HCC, even in association with a high intrahepatic disease burden.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/drug therapy , Fluorouracil/administration & dosage , Liver Neoplasms/drug therapy , Oxaliplatin/administration & dosage , Sorafenib/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , China , Female , Fluorouracil/adverse effects , Hepatic Artery , Humans , Infusions, Intra-Arterial , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Mutation , Oxaliplatin/adverse effects , Progression-Free Survival , Sorafenib/adverse effects , Time Factors
6.
J Immunother Cancer ; 9(6)2021 06.
Article En | MEDLINE | ID: mdl-34168004

BACKGROUND: Neutrophils play a controversial role in tumor development. The function of programmed cell death-1 ligand (PD-L1+) neutrophils, however, may inhibit the cytotoxicity of anti-tumor immunity. In this study, we elucidate the stimulators of PD-L1+ neutrophils in tumor microenvironment (TME) and explore the optimal combination to enhance the effect of lenvatinib by inhibiting PD-L1+ neutrophils in hepatocellular carcinoma. METHODS: Neutrophil infiltration after lenvatinib treatment was examined with RNA sequencing and multicolor flow cytometry analysis in patient samples, subcutaneous and orthotopic mouse models. Neutrophils and T cells were isolated from peripheral blood and tumor tissues and purified with magnetic beads for cytotoxicity assay. Metabolites and cytokines were detected by a biochemical analyzer manufactured by Yellow Springs Instrument (YSI) and proteome profiler cytokines array. In vitro screening of pathway inhibitors was used to identify possible candidates that could reduce PD-L1+ neutrophil infiltration. Further in vivo assays were used for verification. RESULTS: Lenvatinib increased neutrophil recruitment by inducing CXCL2 and CXCL5 secretion in TME. After entering TME, neutrophils polarized toward N2 phenotype. PD-L1 expression was simultaneously upregulated. Thus, lenvatinib efficacy on tumor cells hindered. The increasing PD-L1+ neutrophils positively corelated with a suppressive T cell phenotype. Further investigation indicated that JAK/STAT1 pathway activated by immune-cell-derived interferon γ and MCT1/NF-kB/COX-2 pathway activated by high concentrations of tumor-derived lactate could induce PD-L1+ neutrophils. The latter could be significantly inhibited by COX-2 inhibitor celecoxib. Further in vivo assays verified that Celecoxib decreased the survival of lactate-stimulated PD-L1+ neutrophil and promoted the antitumor effect of lenvatinib. CONCLUSIONS: PD-L1+ neutrophils decrease T cell cytotoxicity. Tumor-derived lactate induces PD-L1 expression on neutrophils via MCT1/NF-κB/COX-2 pathway. Thus, COX-2 inhibitor could reduce PD-L1+ neutrophil and restore T cell cytotoxicity. This may provide a potent addition to lenvatinib.


B7-H1 Antigen/metabolism , Carcinoma, Hepatocellular/drug therapy , Lactic Acid/metabolism , Liver Neoplasms/drug therapy , Neutrophils/metabolism , Phenylurea Compounds/therapeutic use , Quinolines/therapeutic use , Animals , Carcinoma, Hepatocellular/pathology , Female , Humans , Liver Neoplasms/pathology , Mice , Phenylurea Compounds/pharmacology , Quinolines/pharmacology
7.
J Vasc Interv Radiol ; 32(9): 1267-1276.e1, 2021 09.
Article En | MEDLINE | ID: mdl-34166806

PURPOSE: To compare the efficacy and safety of hepatic arterial infusion chemotherapy (HAIC) with a modified fluorouracil, leucovorin, and oxaliplatin (mFOLFOX) regimen with that of transarterial chemoembolization as a locoregional treatment for patients with locally advanced hepatocellular carcinoma (HCC). METHODS: This retrospective study included adult patients with locally advanced HCC who received first-line treatment with either HAIC-mFOLFOX or conventional transarterial chemoembolization monotherapy from January 2015 to December 2016. The outcomes, including tumor response rates, evaluated via imaging assessment using the modified response evaluation criteria in solid tumors; overall survival; progression-free survival; and safety, were compared. The propensity score-matching methodology was used to reduce the influence of confounding factors on the outcomes. RESULTS: The study included 131 patients with locally advanced HCC who underwent transarterial chemoembolization and 101 who received HAIC-mFOLFOX as initial treatment. After propensity score matching (n = 67 in each group), patients who received HAIC-mFOLFOX had a higher objective response rate (43.3% vs 13.4%, P = .001), longer median overall survival (13.9 vs 6.0 months, P < .001), and longer median progression-free survival (6.4 vs 2.8 months, P = .001) than those who underwent transarterial chemoembolization. The survival benefit with HAIC-mFOLFOX was strengthened in patients with HCC with vascular invasion (hazard ratio: 0.379; 95% confidence interval: 0.237-0.607). HAIC-mFOLFOX was associated with lower incidences of severe adverse events (8.9% vs 22.9%) and liver toxicity than transarterial chemoembolization. CONCLUSIONS: Compared with transarterial chemoembolization, HAIC-mFOLFOX is a potentially safer and more effective locoregional therapy for patients with locally advanced HCC.


Carcinoma, Hepatocellular , Chemoembolization, Therapeutic , Liver Neoplasms , Adult , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic/adverse effects , Fluorouracil , Hepatic Artery , Humans , Leucovorin , Liver Neoplasms/therapy , Oxaliplatin , Propensity Score , Retrospective Studies , Treatment Outcome
8.
Immunity ; 54(6): 1304-1319.e9, 2021 06 08.
Article En | MEDLINE | ID: mdl-34048708

Despite mounting evidence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) engagement with immune cells, most express little, if any, of the canonical receptor of SARS-CoV-2, angiotensin-converting enzyme 2 (ACE2). Here, using a myeloid cell receptor-focused ectopic expression screen, we identified several C-type lectins (DC-SIGN, L-SIGN, LSECtin, ASGR1, and CLEC10A) and Tweety family member 2 (TTYH2) as glycan-dependent binding partners of the SARS-CoV-2 spike. Except for TTYH2, these molecules primarily interacted with spike via regions outside of the receptor-binding domain. Single-cell RNA sequencing analysis of pulmonary cells from individuals with coronavirus disease 2019 (COVID-19) indicated predominant expression of these molecules on myeloid cells. Although these receptors do not support active replication of SARS-CoV-2, their engagement with the virus induced robust proinflammatory responses in myeloid cells that correlated with COVID-19 severity. We also generated a bispecific anti-spike nanobody that not only blocked ACE2-mediated infection but also the myeloid receptor-mediated proinflammatory responses. Our findings suggest that SARS-CoV-2-myeloid receptor interactions promote immune hyperactivation, which represents potential targets for COVID-19 therapy.


COVID-19/metabolism , COVID-19/virology , Host-Pathogen Interactions , Lectins, C-Type/metabolism , Membrane Proteins/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasm Proteins/metabolism , SARS-CoV-2/physiology , Angiotensin-Converting Enzyme 2/metabolism , Binding Sites , COVID-19/genetics , Cell Line , Cytokines , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Inflammation Mediators/metabolism , Lectins, C-Type/chemistry , Membrane Proteins/chemistry , Models, Molecular , Neoplasm Proteins/chemistry , Protein Binding , Protein Conformation , Single-Domain Antibodies/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Structure-Activity Relationship
9.
Ann Transl Med ; 9(8): 675, 2021 Apr.
Article En | MEDLINE | ID: mdl-33987373

BACKGROUND: Circular RNAs (circRNAs) are a class of non-coding RNAs that have been demonstrated to play important roles in tumorigenesis. However, how circRNAs regulate the progression of hepatocellular cancer (HCC) remains unclear. METHODS: In the present study, circRNA microarray analyses were performed with HCC tissues to identify circRNAs that are differentially expressed. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis was conducted on HCC cell lines and tissues, and circ0097009 was found to be significantly upregulated. The functions of circ0097009 in HCC were investigated by a series of experiments, including cell proliferation, invasion, and mouse xenograft assays. Additionally, luciferase assays and RNA immunoprecipitation (RIP) assays were used to explore the interactions of circ0097009, microRNA-1261 (miR-1261), and solute carrier family 7 member 11 (SLC7A11) in HCC. RESULTS: Microarray analysis and qRT-PCR verified that circRNA, circ0097009, was significantly upregulated in HCC tissues and cell lines. Knockdown of circ0097009 inhibited the proliferation and invasion of HCC cells. Luciferase reporter assays showed that circ0097009 and SLC7A11 directly bound to miR-1261. Subsequent experiments showed that circ0097009 and SLC7A11 reciprocally regulated their expression via miR-1261 sponging by circ0097009. CONCLUSIONS: Circ0097009 acts as a competing endogenous RNA to regulate the expression of SLC7A11, a key regulator of cancer cell ferroptosis, by sponging miR-1261 in HCC. Circ0097009 may be used as a diagnostic biomarker for HCC and as a potential target for HCC therapy.

10.
Front Oncol ; 10: 580241, 2020.
Article En | MEDLINE | ID: mdl-33163408

Background: The anti-programmed cell death protein-1 (PD-1) inhibitor is one of the second-line therapies for advanced hepatocellular carcinoma (HCC) after sorafenib failure. The goal of this study is to evaluate the feasibility and safety of ablation on the tumor in patients with advanced HCC who had stable disease or atypical response during single anti-PD-1 therapy after sorafenib failure. Atypical response defined as mixed responses in different lesions of the same individual (e.g., active or stable lesions mixed with progressive lesions). Patients and Methods: This proof-of-concept clinical trial enrolled 50 patients treated with an anti-PD-1 inhibitor of nivolumab or pembrolizumab monotherapy between July 2015 and Nov 2017. Thirty-three cases with stable disease or atypical response to anti-PD-1 inhibitor received subtotal thermal ablation. The safety and the response of ablation during anti-PD-1 therapy were evaluated. The survival was estimated by the Kaplan-Meier curve. Results: Of all 50 patients treated with anti-PD-1 therapy, the rate of response, stable disease, atypical and typical progression were 10% (n = 5), 42% (n = 21) 32% (n = 16), and 12% (n = 6), respectively. Additional ablation improved efficacy with tolerable toxicity, and the response rate was increased from 10 to 24% (12/50). The median time to progression, progression-free survival, and overall survival was 6.1 months (95%CI, 2.6-11.2), 5 months (95%CI, 2.9-7.1), and 16.9 months (95%CI, 7.7-26.1), respectively. Conclusions: This proof-of-concept trial suggested that additional ablation may increase the objective response rate with tolerated toxicity and achieved a relatively better median survival, in advanced HCC patients who had stable or atypical progressive diseases during anti-PD-1 therapy, which may provide a potentially promising strategy to treat advanced HCC. Trial registration number: ClinicalTrials.gov identifier: NCT03939975.

11.
Liver Cancer ; 9(3): 338-357, 2020 Jun.
Article En | MEDLINE | ID: mdl-32647635

BACKGROUND AND AIMS: Combining anti-angiogenic therapy with immune checkpoint blockade with anti-programmed cell death-1 (PD-1) antibodies is a promising treatment for hepatocellular carcinoma (HCC). Tyrosine kinase inhibitors are well-known anti-angiogenic agents and offer potential for combination with anti-PD-1 antibodies. This study investigated the possible underlying immunomodulatory mechanisms of combined therapy. METHODS: HCC tissue samples for RNA-sequencing (RNA-seq) were obtained from patients with differential prognoses following anti-PD-1 treatment. Recombinant basic fibroblast growth factor (bFGF) and vascular endothelial growth factor A (VEGFA) were used to stimulate T cells following lenvatinib or sorafenib treatment, respectively. T cell function was analyzed by flow cytometry and lactate dehydrogenase assay. In vivo experiments were conducted in murine H22 and Hepa 1-6 competent models of HCC. Local immune infiltration in the tumor microenvironment (TME) was assessed using multicolor flow cytometry. Gene regulation was evaluated by RNA-seq. Microvascular density was measured by immunohistochemistry, and PD-1 ligand (PD-L1) induction was quantified by western blot. RESULTS: The baseline expression of VEGF and fibroblast growth factor (FGF) in patients with progressive disease was significantly higher than in patients achieving stable disease following anti-PD-1 treatment. VEGFA and bFGF significantly upregulated the expression of PD-1, cytotoxic T-lymphocyte-associated protein-4, and Tim-3 on T cells, while inhibiting the secretion of interferon gamma (IFNG) and granzyme B and suppressing T cell cytotoxicity. This immunosuppressive effect was reverted by lenvatinib but not sorafenib. Furthermore, dual lenvatinib/anti-PD-1 antibody therapy led to better antitumor effects than either sorafenib or fibroblast growth factor receptor (FGFR) inhibitor (BGJ398) in H22 murine models of HCC. Combined lenvatinib/anti-PD-1 treatment also led to long-term immune memory formation, while synergistically modulating the TME and enhancing the cytotoxic effect of T cells. Finally, lenvatinib inhibited PD-L1 expression on human umbilical vein endothelial cells, which improved the function of T cells. CONCLUSIONS: Inhibition of vascular endothelial growth factor receptor and FGFR augmented the efficacy of anti-PD-1 antibodies. Combined lenvatinib/anti-PD-1 treatment appears to exert antitumor activity by synergistically modulating effector T cell function in the TME and by mutually regulating tumor vessel normalization.

12.
Cancer Biol Med ; 17(1): 9-19, 2020 02 15.
Article En | MEDLINE | ID: mdl-32296573

The biological roles of N6 methylation of nucleic acids have been extensively studied. Adenine methylation of RNA is the most prevalent RNA modification and has widespread effects on RNA splicing, translation, localization, and stability. Aberrant dynamic regulation of RNA N6-methyladenosine (m6A) has been reported in numerous human diseases, including several cancers. In recent years, eukaryotic DNA N6-methyladenosine (6mA) has also been reported and implicated in cancer progression and tumorigenesis. In this review, we summarize the contributions of N6-methyladenosine modification to cancer biology and pathogenesis in the context of both RNA and DNA. We also highlight the clinical relevance of targeting these modifications as a therapeutic strategy for cancer.


DNA Methylation/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasms/genetics , RNA Processing, Post-Transcriptional/genetics , Adenosine/analogs & derivatives , Adenosine/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/genetics , DNA Methylation/drug effects , Disease Progression , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/pathology , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/metabolism , RNA Processing, Post-Transcriptional/drug effects , RNA Splicing Factors , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism
13.
Int J Hyperthermia ; 36(1): 1003-1011, 2019.
Article En | MEDLINE | ID: mdl-31544545

Purpose: To evaluate the safety, efficacy, and survival outcomes of computed tomography (CT)-guided thermal ablation for adrenal metastases from hepatocellular carcinoma (HCC). Methods: This long-term retrospective study included 27 male patients (median age, 50 years; range, 34-77 years) with 29 adrenal metastatic tumors associated with HCC who underwent ablation between January 2004 and December 2015. The technical success rate, effectiveness rate, complications, and survival were recorded. Complications were assessed according to the Common Terminology Criteria for Adverse Events. Survival curves were estimated using the Kaplan-Meier method. A Cox regression model was used for the evaluation of factors predicting survival. Results: A total of 33 ablation sessions were performed for the 29 tumors. No ablation-related death was observed, and the incidence of complications was 87.9%. Grade 1-2 complications occurred in 23 of the 33 sessions (69.7%), and grade 3 hypertension was the only major complication, occurring in eight sessions (24.2%). The technical success and effectiveness rates were 93.1% (27 of 29 tumors) and 92.6% (25 of 27 patients), respectively. The median progression-free survival and overall survival (OS) durations for the 27 patients were 6.9 months and 16.8 months, respectively. The median OS duration was longer for patients with adrenal oligometastases (21.8 months) than for those with (12.8 months) multiple metastases (p = .037). Adrenal oligometastases were the only significant predictor of OS (p = .043). Conclusions: CT-guided ablation is a feasible and safe procedure for adrenal metastases from HCC, and it may be more beneficial for patients with adrenal oligometastases.


Adrenal Gland Neoplasms/diagnostic imaging , Adrenal Gland Neoplasms/secondary , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/surgery , Catheter Ablation/methods , Liver Neoplasms/complications , Liver Neoplasms/surgery , Tomography, X-Ray Computed/methods , Adult , Aged , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Retrospective Studies , Survival Analysis
14.
J Vasc Interv Radiol ; 30(3): 349-357.e2, 2019 03.
Article En | MEDLINE | ID: mdl-30819477

PURPOSE: To compare treatment with hepatic arterial infusion of chemotherapy (HAIC) in patients with advanced hepatocellular carcinoma (HCC) with both extrahepatic spread (EHS) and intrahepatic tumor and patients with intrahepatic tumor only. MATERIALS AND METHODS: This single-center retrospective study comprised 116 patients with advanced HCC with both intrahepatic tumor and EHS (EHS group; n = 50) or with intrahepatic tumor only (non-EHS group; n = 66) treated with HAIC including oxaliplatin, fluorouracil, and leucovorin between June 2014 and July 2016. Overall survival (OS) and radiologic responses to treatment were determined and compared between the 2 groups. RESULTS: Both the objective response rate and the clinical benefit rate were higher in the non-EHS group than in the EHS group (37.9% vs 16% objective response rate, P = .010; 81.8% vs 62% clinical benefit rate, P = .017). Median OS was not statistically different between the 2 groups (14.8 months vs 9.8 months, P = .068). Subgroup analysis of OS found that patients with lung metastases survived for a shorter time (OS 7 months) than patients with other metastatic sites (P = .003) and patients free of metastases (P = .001). CONCLUSIONS: HAIC is a potential treatment option for advanced HCC with limited extrahepatic metastases in a population with hepatitis B virus infection.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Fluorouracil/administration & dosage , Hepatic Artery , Leucovorin/administration & dosage , Liver Neoplasms/drug therapy , Oxaliplatin/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/secondary , China , Clinical Decision-Making , Female , Fluorouracil/adverse effects , Humans , Infusions, Intra-Arterial , Leucovorin/adverse effects , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Male , Middle Aged , Oxaliplatin/adverse effects , Patient Selection , Retrospective Studies , Time Factors , Treatment Outcome , Young Adult
15.
Cancer Sci ; 109(12): 3726-3736, 2018 Dec.
Article En | MEDLINE | ID: mdl-30264546

Indoleamine 2,3-dioxygenase 1 (IDO1) is a tryptophan-metabolizing enzyme that is widely distributed in normal or malignant tissues and contributes to immunologic tolerance and immune escape. However, in hepatocellular carcinoma (HCC), the characteristics and mechanism of IDO1 expression have not been well defined. In this study, IDO1 expression in tumor cells (T-IDO1) was frequently detected (109/112) by immunohistochemistry in formalin-fixed paraffin-embedded specimens from HCC patients, and the expression patterns were mostly focal (102/109). Expression of T-IDO1 was significantly associated with the infiltration of CD8+ T cells (P = .043), as well as younger age (<50 years old, P = .02). It was also found that IDO1 had diffuse expression in inflammatory cells in all specimens, which were defined as antigen-presenting cells. Significant correlations among IDO1, IFNG, and CD8A transcriptional levels were observed in freshly resected HCC specimens; moreover, no constitutive IDO1 expression was detected in HCC cell lines until stimulated by interferon-γ through the JAK2-STAT1 signaling pathway, but not type I interferon. Survival analyses showed that increased T-IDO1 and CD8+ T cell infiltration were significantly associated with superior overall survival (OS) (T-IDO1, P = .003; CD8+ T cells, P = .004), and T-IDO1 expression is an independent prognosis factor in both OS and disease-free survival (OS, P = .007; disease-free survival, P = .044). These findings indicated that T-IDO1 expression in HCC is common and is dominantly driven by the host antitumor immune response, which is a favorable prognostic factor in HCC.


CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Hepatocellular/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Liver Neoplasms/metabolism , Adult , Age Factors , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CD8 Antigens/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Interferon-gamma/genetics , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Male , Middle Aged , Prognosis , Survival Analysis , Up-Regulation
16.
Lung Cancer ; 122: 25-31, 2018 08.
Article En | MEDLINE | ID: mdl-30032841

OBJECTIVE: To assess the safety and effectiveness of computed-tomography-guided (CT-guided) percutaneous radiofrequency ablation (RFA) for lung neoplasms adjacent to the pericardium. MATERIALS AND METHODS: This retrospective study was approved by the institutional ethics committee, and all patients provided written informed consent. Between January 1, 2012 and March 31, 2016, 49 consecutive patients (32 men and 17 women; mean age 47.9 years; range 16-67 years) with 51 tumors (mean diameter 17.7 ±â€¯7.2 mm; range 4-30 mm) located within 10 mm of the pericardium underwent percutaneous CT-guided RFA. The technical success rate, local progression-free survival (LPFS), risk factors for local progression, and safety of this technique were evaluated. RESULTS: The technical success rate was 96.1% (49/51), and the LPFS was 98.0%, 93.0%, 84.0%, and 77.0% at 3, 6, 12, and 24 months, respectively. A tumor size >2.0 cm (p = .045) and the primary lung tumor types (p = .013) were associated with local progression. No treatment-related deaths occurred. The incidence of major complications was 15.7%; complications included pneumothorax (5.9%, 3/51), pleural effusion (3.9%, 2/51), pneumonitis (3.9%, 2/51), and hemothorax (2.0%, 1/51). CONCLUSION: Percutaneous CT-guided RFA can be a safe and effective option for the treatment of lung malignancies adjacent to the pericardium.


Lung Neoplasms/surgery , Lung/pathology , Pericardium/pathology , Radiofrequency Ablation , Adolescent , Adult , Aged , Female , Humans , Lung/surgery , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Pericardium/surgery , Retrospective Studies , Survival Analysis , Tomography, X-Ray Computed , Young Adult
17.
Mol Med Rep ; 17(5): 7319-7324, 2018 05.
Article En | MEDLINE | ID: mdl-29568951

To investigate the effect of liposome Lipofectamine® 2000­mediated HSP27 plasmid transfection in A549 human alveolar type II epithelial cell line on collagen synthesis during transforming growth factor­ß1 (TGF­ß1)­induced type II epithelial cell transition to myofibroblasts. Cells were transfected with varying ratios of the Lipofectamine® 2000­mediated heat shock protein 27 (HSP27) plasmid and the transfection efficiency was determined using flow cytometry. The maximum transfection efficacy was confirmed by laser confocal microscopy. HSP gene expression and the most efficient HSP27 plasmid were determined using reverse transcription­quantitative polymerase chain reaction. Western blot analysis was used to examine HSP27 and collagen expression levels. With a transfection efficiency of 83%, the 8 µg:20 µl ratio of liposome: Plasmid had the highest transfection levels. Among the four different interference sequences in the HSP27 plasmid, the D sequence had the highest interference effect with 70% silencing of the HSP27 gene. The expression of type I and III collagen in TGF­ß1­induced transition of A549 human alveolar type II epithelial cell line to myofibroblasts was significantly downregulated by the successful transfection with HSP27­interfering plasmid. The expression of type I and III collagen in the TGF­ß1­induced transition of A549 cells to myofibroblasts was significantly downregulated by transfection of A549 cells with HSP27 plasmid D­interfering sequence and optimal ratio of Lipofectamine® 2000 and HSP27 plasmid.


Alveolar Epithelial Cells/metabolism , Collagen/metabolism , HSP27 Heat-Shock Proteins/genetics , Lipids/chemistry , Liposomes/chemistry , Transfection/methods , A549 Cells , Biosynthetic Pathways , Collagen/analysis , HSP27 Heat-Shock Proteins/analysis , Heat-Shock Proteins , Humans , Molecular Chaperones , Plasmids/administration & dosage , Plasmids/genetics
18.
J Hepatol ; 69(1): 60-69, 2018 07.
Article En | MEDLINE | ID: mdl-29471013

BACKGROUND & AIMS: To compare the overall survival (OS) and disease progression free survival (PFS) in patients with advanced hepatocellular carcinoma (Ad-HCC) who are undergoing hepatic arterial infusion (HAI) of oxaliplatin, fluorouracil/leucovorin (FOLFOX) treatment vs. sorafenib. METHODS: This retrospective study was approved by the ethical review committee, and informed consent was obtained from all patients before treatment. HAI of FOLFOX (HAIF) was recommended as an alternative treatment option for patients who refused sorafenib. Of the 412 patients with Ad-HCC (376 men and 36 women) between Jan 2012 to Dec 2015, 232 patients were treated with sorafenib; 180 patients were given HAIF therapy. The median age was 51 years (range, 16-82 years). Propensity-score matched estimates were used to reduce bias when evaluating survival. Survival curves were calculated by performing the Kaplan-Meier method and compared by using the log-rank test and Cox regression models. RESULTS: The median PFS and OS in the HAIF group were significantly longer than those in the sorafenib group (PFS 7.1 vs. 3.3 months [RECIST]/7.4 vs. 3.6 months [mRECIST], respectively; OS 14.5 vs. 7.0 months; p <0.001 for each). In the propensity-score matched cohorts (147 pairs), both PFS and OS in the HAIF group were longer than those in the sorafenib group (p <0.001). At multivariate analysis, HAIF treatment was an independent factor for PFS (hazard ratio [HR] 0.389 [RECIST]/0.402 [mRECIST]; p <0.001 for each) and OS (HR 0.129; p <0.001). CONCLUSION: HAIF therapy may improve survival compared to sorafenib in patients with Ad-HCC. A prospective randomized trial is ongoing to confirm this finding. LAY SUMMARY: We compared the hepatic arterial infusion of FOLFOX (a combination chemotherapy) with sorafenib (a tyrosine kinase inhibitor) in patients with advanced hepatocellular carcinoma, retrospectively. It was found that hepatic arterial infusion of FOLFOX therapy may improve both progression free and overall survival in patients with advanced hepatocellular carcinoma.


Carcinoma, Hepatocellular/drug therapy , Fluorouracil/administration & dosage , Leucovorin/administration & dosage , Liver Neoplasms/drug therapy , Neoplasm Staging , Oxaliplatin/administration & dosage , Sorafenib/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/mortality , China/epidemiology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Follow-Up Studies , Hepatic Artery , Humans , Immunosuppressive Agents/administration & dosage , Infusions, Intra-Arterial , Liver Neoplasms/diagnosis , Liver Neoplasms/mortality , Male , Middle Aged , Retrospective Studies , Survival Rate/trends , Treatment Outcome , Vitamin B Complex/administration & dosage , Young Adult
19.
Int J Hyperthermia ; 34(1): 59-67, 2018 02.
Article En | MEDLINE | ID: mdl-28540809

BACKGROUND: The aim of this study was to evaluate the therapeutic outcome of percutaneous computed tomography (CT)-guided radiofrequency ablation (RFA) for extrahepatic oligometastases of hepatocellular carcinoma (HCC). METHODS: Institutional review board approval was obtained for this retrospective study, and all patients provided written informed consent. Between April 2004 and December 2015, 116 oligometastases (diameter, 5-50 mm; 20.3 ± 10.4) in 79 consecutive HCC patients (73 men and 6 women; average age, 50.3 years ±13.0) were treated with RFA. We focussed on patients with 1-3 extrahepatic metastases (EHM) confined to 1-2 organs (including the lung, adrenal gland, bone, lymph node and pleura/peritoneum) who were treated naïve with curative intent. Survival, technical success and safety were evaluated. The log-rank test and Cox proportional hazards regression models were used to analyse the survival data. RESULTS: No immediate technical failure occurred, and at 1 month, the technique effectiveness rate was determined to be 95.8%. After a median follow-up time of 28.0 months (range, 6-108 months), the 1-, 2- and 3-year overall survival (OS) rates were 91, 70 and 48%, respectively, with a median survival time of 33.5 months. Time to unoligometastatic progression (TTUP) of less than 6 months (p < 0.001) and a Child-Pugh score of more than 5 (p = 0.001) were significant indicators of shorter OS. The 1-, 2- and 3-year disease free survival (DFS) rates were 34, 21 and 8%, respectively, with a median DFS time of 6.8 months. DFS was better for those with lung metastases (p = 0.006). Major complication occurred in nine (9.5%, 9/95) RFA sessions without treatment-related mortality. CONCLUSIONS: CT-guided RFA for oligometastatic HCC may provide favourable efficacy and technical success with a minimally invasive approach.


Carcinoma, Hepatocellular/surgery , Liver Neoplasms/surgery , Radiofrequency Ablation/methods , Tomography, X-Ray Computed/methods , Carcinoma, Hepatocellular/pathology , Female , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Retrospective Studies , Survival Rate , Treatment Outcome
20.
J Cancer ; 8(17): 3506-3513, 2017.
Article En | MEDLINE | ID: mdl-29151935

Background & Aims: We compared the efficacy of transcatheter arterial chemoembolization (TACE) in combination with CT-guided radiofrequency ablation (RFA) with that of surgical resection (SR) in patients with hepatocellular carcinoma (HCC) within the up-to-seven criteria. Methods: From January 2004 to December 2014, 420 multicenter consecutive patients with HCC who conformed to the up-to-seven criteria and initially received either TACE plus CT-guided RFA (TACE-RFA) or SR were enrolled. A matched cohort composed of 206 patients was selected after adjustment with propensity score matching. The overall survival (OS) of each patient was calculated with the Kaplan-Meier method and compared by the log-rank test. Results: The median OS and 1-, 3-, and 5-year survival rates were 56.0 months, 96.1%, 76.7% and 41.3% in the TACE-RFA group and 58.0 months, 96.1%, 86.4% and 46.2% in the SR group, respectively. There was no significant difference in OS between the two groups (P = 0.138). For patients with HCC beyond the Milan criteria, TACE-RFA provided a longer median OS than SR (52.0 vs 45.0 months, P = 0.023). Conclusions: Treatment by TACE-RFA conferred an OS rate comparable with that of SR in patients within the up-to-seven criteria. For patients with HCC between the Milan and the up-to-seven criteria, TACE-RFA might be superior to SR for survival prolongation.

...