Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Phytomedicine ; 131: 155790, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38851099

ABSTRACT

BACKGROUND: A balanced protein homeostasis network helps cholangiocarcinoma (CCA) maintain their oncogenic growth, and disrupting proteostasis therapeutically will induce proteotoxic stress. Phosphatase and tensin homolog (PTEN) have been reported to be involved in proteostasis, and PTEN-associated pathways are commonly altered in CCA. Celastrol, a triterpene from plants, exhibits cytotoxic effects in various types of cancer. However, the underlying mechanisms remain unclear. PURPOSE: We investigated the therapeutic effect of celastrol in CCA and identified the molecular characteristics of tumors that were sensitive to celastrol. The target of celastrol was explored. We then evaluated the candidate combination therapeutic strategy to increase the effectiveness of celastrol in celastrol-insensitive CCA tumors. METHODS: Various CCA cells were categorized as either celastrol-sensitive or celastrol-insensitive based on their response to celastrol. The molecular characteristics of cells from different groups were determined by RNA-seq. PTEN status and its role in proteasome activity in CCA cells were investigated. The CMAP analysis, molecular docking, and functional assay were performed to explore the effect of celastrol on proteasome activities. The correlation between PTEN status and clinical outcomes, as well as proteasomal activity, were measured in CCA patients. The synergistic therapeutic effect of autophagy inhibitors on celastrol-insensitive CCA cells were measured. RESULTS: Diverse responses to celastrol were observed in CCA cells. PTEN expression varied among different CCA cells, and its status could impact cell sensitivity to celastrol: PTENhigh tumor cells were resistant to celastrol, while PTENlow cells were more sensitive. Celastrol induced proteasomal dysregulation in CCA cells by directly targeting PSMB5. Cells with low PTEN status transcriptionally promoted proteasome subunit expression in an AKT-dependent manner, making these cells more reliant on proteasomal activities to maintain proteostasis. This caused the PTENlow CCA cells sensitive to celastrol. A negative correlation was found between PTEN levels and the proteasome signature in CCA patients. Moreover, celastrol treatment could induce autophagy in PTENhigh CCA cells. Disrupting the autophagic pathway in PTENhigh CCA cells enhanced the cytotoxic effect of celastrol. CONCLUSION: PTEN status in CCA cells determines their sensitivity to celastrol, and autophagy inhibitors could enhance the anti-tumor effect in PTENhigh CCA.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , PTEN Phosphohydrolase , Pentacyclic Triterpenes , Triterpenes , Cholangiocarcinoma/drug therapy , Pentacyclic Triterpenes/pharmacology , PTEN Phosphohydrolase/metabolism , Humans , Cell Line, Tumor , Bile Duct Neoplasms/drug therapy , Triterpenes/pharmacology , Molecular Docking Simulation , Tripterygium/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/drug effects , Autophagy/drug effects , Bortezomib/pharmacology
2.
Cell Death Dis ; 15(4): 300, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684648

ABSTRACT

The treatment of hepatocellular carcinoma (HCC) is particularly challenging due to the inherent tumoral heterogeneity and easy resistance towards chemotherapy and immunotherapy. Arsenic trioxide (ATO) has emerged as a cytotoxic agent effective for treating solid tumors, including advanced HCC. However, its effectiveness in HCC treatment remains limited, and the underlying mechanisms are still uncertain. Therefore, this study aimed to characterize the effects and mechanisms of ATO in HCC. By evaluating the susceptibilities of human and murine HCC cell lines to ATO treatment, we discovered that HCC cells exhibited a range of sensitivity to ATO treatment, highlighting their inherent heterogeneity. A gene signature comprising 265 genes was identified to distinguish ATO-sensitive from ATO-insensitive cells. According to this signature, HCC patients have also been classified and exhibited differential features of ATO response. Our results showed that ATO treatment induced reactive oxygen species (ROS) accumulation and the activation of multiple cell death modalities, including necroptosis and ferroptosis, in ATO-sensitive HCC cells. Meanwhile, elevated tumoral immunogenicity was also observed in ATO-sensitive HCC cells. Similar effects were not observed in ATO-insensitive cells. We reported that ATO treatment induced mitochondrial injury and mtDNA release into the cytoplasm in ATO-sensitive HCC tumors. This subsequently activated the cGAS-STING-IFN axis, facilitating CD8+ T cell infiltration and activation. However, we found that the IFN pathway also induced tumoral PD-L1 expression, potentially antagonizing ATO-mediated immune attack. Additional anti-PD1 therapy promoted the anti-tumor response of ATO in ATO-sensitive HCC tumors. In summary, our data indicate that heterogeneous ATO responses exist in HCC tumors, and ATO treatment significantly induces immunogenic cell death (ICD) and activates the tumor-derived mtDNA-STING-IFN axis. These findings may offer a new perspective on the clinical treatment of HCC and warrant further study.


Subject(s)
Arsenic Trioxide , Carcinoma, Hepatocellular , Immunogenic Cell Death , Liver Neoplasms , Membrane Proteins , Nucleotidyltransferases , Arsenic Trioxide/pharmacology , Arsenic Trioxide/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/immunology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Humans , Animals , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Immunogenic Cell Death/drug effects , Cell Line, Tumor , Interferons/metabolism , Signal Transduction/drug effects , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL
3.
Sci Transl Med ; 15(704): eadd7464, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37437018

ABSTRACT

Gemcitabine is a nucleoside analog that has been successfully used in the treatment of multiple cancers. However, intrinsic or acquired resistance reduces the chemotherapeutic potential of gemcitabine. Here, we revealed a previously unappreciated mechanism by which phosphatase and tensin homolog (PTEN), one of the most frequently mutated genes in human cancers, dominates the decision-making process that is central to the regulation of gemcitabine efficacy in cholangiocarcinoma (CCA). By investigating a gemcitabine-treated CCA cohort, we found that PTEN deficiency was correlated with the improved efficacy of gemcitabine-based chemotherapy. Using cell-based drug sensitivity assays, cell line-derived xenograft, and patient-derived xenograft models, we further confirmed that PTEN deficiency or genetic-engineering down-regulation of PTEN facilitated gemcitabine efficacy both in vitro and in vivo. Mechanistically, PTEN directly binds to and dephosphorylates the C terminus of the catalytic subunit of protein phosphatase 2A (PP2Ac) to increase its enzymatic activity, which further dephosphorylates deoxycytidine kinase (DCK) at Ser74 to diminish gemcitabine efficacy. Therefore, PTEN deficiency and high phosphorylation of DCK predict a better response to gemcitabine-based chemotherapy in CCA. We speculate that the combination of PP2A inhibitor and gemcitabine in PTEN-positive tumors could avoid the resistance of gemcitabine, which would benefit a large population of patients with cancer receiving gemcitabine or other nucleoside analogs.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Phosphorylation , Gemcitabine , Nucleosides , Bile Ducts, Intrahepatic , PTEN Phosphohydrolase
4.
Gastroenterology ; 164(3): 424-438, 2023 03.
Article in English | MEDLINE | ID: mdl-36436593

ABSTRACT

BACKGROUND & AIMS: In eukaryotes, the ubiquitin-proteasome system and the autophagy-lysosome pathway are essential for maintaining cellular proteostasis and associated with cancer progression. Our previous studies have demonstrated that phosphatase and tensin homolog (PTEN), one of the most frequently mutated genes in human cancers, limits proteasome abundance and determines chemosensitivity to proteasome inhibitors in cholangiocarcinoma (CCA). However, whether PTEN regulates the lysosome pathway remains unclear. METHODS: We tested the effects of PTEN on lysosome biogenesis and exosome secretion using loss- and gain-of-function strategies in CCA cell lines. Using in vitro dephosphorylation assays, we explored the regulatory mechanism between PTEN and the key regulator of lysosome biogenesis, transcription factor EB (TFEB). Using the migration assays, invasion assays, and trans-splenic liver metastasis mouse models, we evaluated the function of PTEN deficiency, TFEB-mediated lysosome biogenesis, and exosome secretion on tumor metastasis. Moreover, we investigated the clinical significance of PTEN expression and exosome secretion by retrospective analysis. RESULTS: PTEN facilitated lysosome biogenesis and acidification through its protein phosphatase activity to dephosphorylate TFEB at Ser211. Notably, PTEN deficiency increased exosome secretion by reducing lysosome-mediated degradation of multi-vesicular bodies, which further facilitated the proliferation and invasion of CCA. TFEB agonist curcumin analog C1 restrained the metastatic phenotype caused by PTEN deficiency in mouse models, and we highlighted the correlation between PTEN deficiency and exosome secretion in clinical cohorts. CONCLUSIONS: In CCA, PTEN deficiency impairs lysosome biogenesis to facilitate exosome secretion and cancer metastasis in a TFEB phosphorylation-dependent manner.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cholangiocarcinoma , Exosomes , PTEN Phosphohydrolase , Animals , Humans , Mice , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cholangiocarcinoma/metabolism , Disease Models, Animal , Exosomes/metabolism , Lysosomes/physiology , Proteasome Endopeptidase Complex , PTEN Phosphohydrolase/metabolism , Retrospective Studies
5.
Cancer Sci ; 113(12): 4151-4164, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36114745

ABSTRACT

Oncogene-derived metabolic reprogramming is important for anabolic growth of cancer cells, which is now considered to be not simply rely on glycolysis. Pentose phosphate pathway and tricarboxylic acid cycle also play pivotal roles in helping cancer cells to meet their anabolic and energy demands. The present work focused on gankyrin, a relatively specific oncogene in hepatocellular carcinoma (HCC), and its impact on glycolysis and mitochondrial homeostasis. Metabolomics, RNA-seq analysis, and subsequent conjoint analysis illustrated that gankyrin regulated the pentose phosphate pathway (PPP), tricarboxylic acid (TCA) cycle, and mitochondrial function and homeostasis, which play pivotal roles in tumor development. Mechanistically, gankyrin was found to modulate HCC metabolic reprogramming via TIGAR. Gankyrin positively regulated the transcription of TIGAR through Nrf2, which bound to the antioxidant response elements (AREs) in the promoter of TIGAR. Interestingly, TIGAR feedback regulated the transcription of Nrf2 and subsequently gankyrin by promoting nuclear importation of PGC1α. The loop between gankyrin, Nrf2, and TIGAR accelerated glucose metabolism toward the PPP and TCA cycle, which provided vital building blocks, such as NADPH, ATP, and ribose of tumor and further facilitated the progression of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Citric Acid Cycle , Liver Neoplasms/pathology , Glycolysis , Glucose/metabolism
6.
Front Immunol ; 13: 943066, 2022.
Article in English | MEDLINE | ID: mdl-36159865

ABSTRACT

Background: Immune checkpoint inhibitor (ICI)-combined chemotherapy in advanced intrahepatic cholangiocarcinoma has been proved to have more efficacy in a series of clinical trials. However, whether the tumor microenvironment (TME) plays a vital role in immune-combined therapy has not been rigorously evaluated. Methods: Firstly, we assayed the immunogenic properties of GEM-based chemotherapy. Then, 12 ICC patients treated with PD-1 inhibitor (sintilimab) combined with gemcitabine and cisplatin (GemCis) from a phase 2 clinical trial (ChiCTR2000036652) were included and their immune-related gene expression profiles were analyzed using RNA from baseline tumor samples. Immune-related signature correlating with clinical outcome was identified according to the 12 ICC patients, and its predictive value was validated in an ICC cohort with 26 patients. Multiplexed immunofluorescence (mIF) and flow cytometry (FCM) analysis were performed to evaluate the immune-related molecules with therapeutic outcomes. Results: GEM-based chemotherapy induced immunogenic cell death of cholangiocarcinoma cells, together with increased CD274 expression. In an ICC cohort, we found that upregulation of immune-checkpoint molecules and immune response-related pathways were significantly related to better clinical outcome. On the contrary, baseline immune-cell proportions in tumor tissues did not show any correlation with clinical benefit between responders and non-responders. Immune-related signature (including six genes) correlating with clinical outcome was identified according to the 12 ICC patients, and its predictive value was validated in a small ICC cohort with 26 patients. Conclusion: Immune-related RNA signature predicts the outcome of PD-1 inhibitor-combined GEMCIS therapy in advanced intrahepatic cholangiocarcinoma, which could be tested as a biomarker for immune-chemotherapy in the future.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic/metabolism , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Cisplatin/therapeutic use , Clinical Trials, Phase II as Topic , Humans , Immune Checkpoint Inhibitors/therapeutic use , RNA , Tumor Microenvironment
7.
Cell Rep ; 39(12): 110987, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35732131

ABSTRACT

The liver is one of the most-favored distant metastatic sites for solid tumors, and interactions between cancer cells and components of the hepatic microenvironment are essential for liver metastasis (LM). Although sex is one of the determinants for primary liver cancer, sexual dimorphism in LM (SDLM) and the underlying mechanisms remain unclear. We herein demonstrate a significant male-biased SDLM, which is attributed to host androgen/androgen receptor (Ar) signaling that promotes hepatic seeding of tumor cells and subsequent outgrowth in a neutrophil-dependent manner. Mechanistically, androgen/Ar signaling promotes hepatic accumulation of neutrophils by promoting proliferation and development of neutrophil precursors in the bone marrow, as well as modulating hepatic recruitment of neutrophils and their functions. Antagonizing the androgen/Ar/neutrophil axis significantly mitigates LM in males. Our data thus reveal an important role of androgen in LM and suggest that androgen/Ar modulation represents a promising target for LM therapy in men.


Subject(s)
Androgens , Liver Neoplasms , Neutrophils , Sex Characteristics , Humans , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Male , Neutrophils/cytology , Receptors, Androgen , Tumor Microenvironment
8.
Cancer Lett ; 501: 187-199, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33220333

ABSTRACT

Gallbladder cancer (GBC) is an aggressive malignancy of biliary tract with poor prognosis. Although several studies have shown the frequency of relevant genetic alterations, there are few genetic models or translational studies that really benefit for GBC treatment in the era of precision medicine. By targeted sequencing and immunohistochemistry staining, we identified that phosphate and tension homology deleted on chromosome ten (PTEN) was frequently altered in GBC specimens, and loss of PTEN expression was independently correlated with poor survival outcomes. Further drug screening assays revealed proteasome inhibitor bortezomib as a promising agent for GBC treatment, and knockdown of PTEN increased bortezomib efficacy both in vivo and in vitro. Therapeutic evaluation of patient derived xenografts (PDXs) strongly supported the utilization of bortezomib in PTEN deficient GBC. Mechanically, functional PTEN inhibited ARE-dependent transcriptional activity, the same machinery regulating the transcription of proteasome subunits, thus PTEN suppressed proteasome activity and bortezomib sensitivity. Through siRNA screening, we identified the ARE-related transcriptional suppressor BACH1 involved in PTEN-mediated proteasome inhibition and regulated by PTEN-AKT1 axis. In summary, our study indicates that proteasome activity represents a prime therapeutic target in PTEN-deficient GBC tumors, which is worthy of further clinical validation.


Subject(s)
Bortezomib/administration & dosage , Down-Regulation , Gallbladder Neoplasms/drug therapy , Mutation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Adult , Aged , Animals , Bortezomib/pharmacology , Cell Line, Tumor , Female , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Mice , Middle Aged , Proteasome Endopeptidase Complex/metabolism , Survival Analysis , Xenograft Model Antitumor Assays , Young Adult
9.
Sci Transl Med ; 12(562)2020 09 23.
Article in English | MEDLINE | ID: mdl-32967970

ABSTRACT

Patient-derived xenografts (PDXs) and PDX-derived cells (PDCs) are useful in preclinical research. We performed a drug screening assay using PDCs and identified proteasome inhibitors as promising drugs for cholangiocarcinoma (CCA) treatment. Furthermore, we determined that phosphate and tensin homology deleted on chromosome ten (PTEN) deficiency promotes protein synthesis and proteasome subunit expression and proteolytic activity, creating a dependency on the proteasome for cancer cell growth and survival. Thus, targeting the proteasome machinery with the inhibitor bortezomib inhibited the proliferation and survival of CCA cells lacking functional PTEN. Therapeutic evaluation of PDXs, autochthonous mouse models, and patients confirmed this dependency on the proteasome. Mechanistically, we found that PTEN promoted the nuclear translocation of FOXO1, resulting in the increased expression of BACH1 and MAFF BACH1 and MAFF are transcriptional regulators that recognize the antioxidant response element, which is present in genes encoding proteasome subunits. PTEN induced the accumulation and nuclear translocation of these proteins, which directly repressed the transcription of genes encoding proteasome subunits. We revealed that the PTEN-proteasome axis is a potential target for therapy in PTEN-deficient CCA and other PTEN-deficient cancers.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Animals , Bile Duct Neoplasms/drug therapy , Bile Ducts, Intrahepatic , Bortezomib/pharmacology , Bortezomib/therapeutic use , Cell Line, Tumor , Cholangiocarcinoma/drug therapy , Humans , Mice , PTEN Phosphohydrolase/genetics , Proteasome Endopeptidase Complex
10.
Hepatology ; 71(6): 2005-2022, 2020 06.
Article in English | MEDLINE | ID: mdl-31541481

ABSTRACT

BACKGROUND AND AIMS: Cancer cell survival depends on the balance between reactive oxygen species production and scavenging, which is regulated primarily by NRF2 during tumorigenesis. Here, we demonstrate that deletion of RBP5-mediating protein (RMP) in an autonomous mouse model of intrahepatic cholangiocarcinoma (ICC) delays tumor progression. APPROACH AND RESULTS: RMP-overexpressing tumor cells exhibited enhanced tolerance to oxidative stress and apoptosis. Mechanistically, RMP competes with NRF2 for binding to the Kelch domain of KEAP1 (Kelch-like ECH-associated protein 1) through the E**E motif, leading to decreased NRF2 degradation via ubiquitination, thus increasing NRF2 nuclear translocation and downstream transactivation of antioxidant genes. This RMP-KEAP1-NRF2 axis promotes ICC tumorigenesis, metastasis, and drug resistance. Consistent with these findings, the RMP level in human ICC is positively correlated with the protein level of NRF2 and is associated with poor prognosis. CONCLUSION: These findings reveal that RMP is involved in the oxidative stress defense program and could be exploited for targeted cancer therapies.


Subject(s)
Carcinogenesis , Cholangiocarcinoma/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Repressor Proteins/metabolism , Animals , Apoptosis , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Cell Line , Cell Transformation, Neoplastic/metabolism , Cholangiocarcinoma/pathology , Drug Resistance, Neoplasm , Humans , Mice , Oxidative Stress
11.
Strahlenther Onkol ; 196(3): 270-279, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31748837

ABSTRACT

PURPOSE: The efficacy and tolerability of adding chemotherapy to radiotherapy in the era of intensity-modulated radiation therapy (IMRT) remain controversial among older patients with nasopharyngeal carcinoma (NPC). The present study compared IMRT alone with IMRT in combination with chemotherapy in elderly NPC patients. METHODS: Between January 2011 and December 2014, 102 patients aged >65 years with NPC who received IMRT alone (IMRT group) or IMRT in combination with chemotherapy (IMRT/CT group) were enrolled. Patients from both treatment arms were pair-matched (1:1 ratio) based on six clinical factors. Differences in overall survival (OS), disease-free survival (DFS), locoregional relapse-free survival (LRRFS), and distant metastasis-free survival (DMFS) were assessed using the Kaplan-Meier method and Cox proportional hazards models, whereas the toxicity profile was assessed using Common Terminology Criteria for Adverse Events (CTCAE) version 4. RESULTS: No significant differences were noted in OS (72.1% vs. 72.5%, p = 0.799), DFS (65.9% vs. 70.1%, p = 0.733), LRRFS (76.4% vs. 71.6%, p = 0.184), and DMFS (90.8% vs. 98.0%, p = 0.610) between the IMRT and IMRT/CT groups. Multivariate analyses showed that chemotherapy was not an independent factor for OS, DFS, LRRFS, and DMFS. However, the incidences of grade 3 vomiting/nausea (p = 0.000), leukopenia/neutropenia (p = 0.000), thrombocytopenia (p = 0.041), and anemia (p = 0.040) were significantly higher in the IMRT/CT group compared with the IMRT group. No grade 4 toxicities were observed. CONCLUSION: IMRT alone was similar to IMRT/CT in treating elderly NPC patients (age >65 years), with comparable survival outcomes and less grade 3 toxicities.


Subject(s)
Chemoradiotherapy , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/therapy , Radiotherapy, Intensity-Modulated , Aged , Chemoradiotherapy/methods , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Proportional Hazards Models , Radiotherapy, Intensity-Modulated/methods , Retrospective Studies
12.
Cancer Lett ; 421: 161-169, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29452147

ABSTRACT

Kras mutations are among the most common genetic abnormalities in human neoplasms, including cholangiocarcinomas, pancreatic cancer and colon cancer. PTEN has previously been associated with cholangiocarcinoma development in murine models. Here, we have established novel mouse models of neoplasms by liver-specific and biliary-pancreatic Kras activation and PTEN deletion. By liver-specific disruption of PTEN and activation of Kras in mice caused rapid development of intrahepatic biliary epithelial proliferative lesions (Intrahepatic cholangiocarcinoma, ICC), which progress through dysplasia to invasive carcinoma. In contrast, Kras activation in combination with heterozygous PTEN deletion induced mixed carcinomas of liver (both ICC and hepatocellular carcinoma, HCC), whereas Kras activation alone did not induce biliary tract neoplasm. Use of Sox9-Cre-LoxP-based approach to coordinately delete PTEN and activate Kras in the adult mouse resulted in not only development of low-grade biliary lesions (ICC and extrahepatic bile duct carcinoma, ECC) but also pancreatic carcinomas. Our data provide a functional link between PTEN gene status, hepatobiliary cell fate, and HCC, biliary carcinoma, pancreatic cancer pathogenesis, and present novel genetically engineered mouse models of PTEN loss-driven malignancy.


Subject(s)
Gene Deletion , Genes, ras , Liver Neoplasms, Experimental/pathology , PTEN Phosphohydrolase/genetics , Pancreatic Neoplasms/pathology , Animals , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Mice , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Transgenes
13.
Oral Oncol ; 74: 34-39, 2017 11.
Article in English | MEDLINE | ID: mdl-29103749

ABSTRACT

OBJECTIVES: The efficacy of various chemotherapy regimens in nasopharyngeal carcinoma (NPC) remains under debate. We compared the efficacy and toxicity of a taxane-based regimen and regimen including fluorouracil in NPC. MATERIALS AND METHODS: Eight-hundred and six patients with stage II-IVB NPC from four institutions in China were pair-matched (1:1 ratio) to the cisplatin plus fluorouracil (PF) group or cisplatin plus taxanes (TP) group using eight clinical factors. Overall survival (OS), disease-free survival (DFS), locoregional relapse-free survival (LRRFS) and distant metastasis-free survival (DMFS) were assessed using the Kaplan-Meier method and Cox regression model. Toxicities were assessed in all patients. RESULTS: Three-year DFS was significantly better in the TP group than PF group (82.5% vs. 72.7%, P=0.002), with no significant difference in OS, LRRFS or DMFS. TP led to significantly better DFS compared to PF in the subgroups advanced stage NPC, patients aged ≤45-years-old and female patients. In multivariate analysis, chemotherapy regimen was an independent prognostic factor for DFS [hazard ratio, 0.591, 95% CI 0.444-0.786, P=0.000]. Grade 3-4 leukopenia, neutropenia and anemia were significantly more common in the TP group; grade 3-4 mucositis, vomiting, vasculitis and diarrhea were more common in the PF group. CONCLUSION: Taxane-based regimens have a higher efficacy in NPC than regimens including fluorouracil, especially in patients with advanced stage, patients aged≤45-years-old and female patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/drug therapy , Fluorouracil/administration & dosage , Nasopharyngeal Neoplasms/drug therapy , Taxoids/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma/pathology , Female , Humans , Male , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Retrospective Studies , Survival Analysis , Treatment Outcome
14.
Oncotarget ; 8(8): 14078-14088, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-27845907

ABSTRACT

BACKGROUND: The relative efficacy of induction chemotherapy (IC) followed by concurrent chemoradiotherapy (CCRT) versus IC followed by radiotherapy (RT) alone in locoregionally advanced NPC remains unclear. METHODS: A total of 877 patients with locally advanced NPC who underwent IC/CCRT or IC/RT at four institutions in China between January 2004 and December 2010 were retrospectively assessed. IC was cisplatin-based combination chemotherapy; concurrent chemotherapy, single agent cisplatin. Two-dimensional conventional radiotherapy (2DCRT) was the radiotherapy technique. All patients were matched in an equal ratio using a pair-matched method. Overall survival (OS), disease-free survival (DFS), distant metastasis-free survival (DMFS), locoregional relapse-free survival (LRRFS) and toxicities were assessed. RESULTS: Eligible patients were matched (n = 642; 321 patients per arm) based on eight clinicopathological characteristics. Five-year OS, DFS, DMFS, and LRRFS were 76%, 70%, 86%, and 88% for IC/CCRT and 75%, 70%, 90%, and 91% for IC/RT, respectively. There were no statistically significant survival differences between arms (P>0.05), even in subgroup analysis. In multivariate analysis, treatment (IC/CCRT vs. IC/RT) was not an independent prognostic factor for any survival end-point. Grade 3/4 acute gastrointestinal toxicities (vomiting, nausea) and hematological toxicities (leucopenia/neutropenia, thrombocytopenia and anemia) were significantly more common in the IC/CCRT arm than IC/RT arm during RT. CONCLUSION: Overall, IC/CCRT failed to demonstrate any survival advantage but higher acute toxicities over IC/RT in locoregionally advanced NPC.


Subject(s)
Carcinoma/drug therapy , Carcinoma/radiotherapy , Chemoradiotherapy/methods , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/radiotherapy , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma/mortality , Cisplatin/administration & dosage , Cisplatin/adverse effects , Disease-Free Survival , Female , Humans , Induction Chemotherapy/methods , Kaplan-Meier Estimate , Male , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/mortality , Prognosis , Retrospective Studies , Treatment Outcome , Young Adult
15.
Adv Mater ; 29(5)2017 Feb.
Article in English | MEDLINE | ID: mdl-27869342

ABSTRACT

A 2D peptidosheet unravels CD47 as a potential biomarker to image hepatocarcinoma and cholangiocarcinoma cells and tissues. Supramolecular assembly between water-soluble 2D MoS2 and a peptide probe produces the 2D peptidosheet suited for the profiling of hepatocarcinoma and cholangiocarcinoma tissues over healthy tissues on clinical specimens.


Subject(s)
CD47 Antigen/chemistry , Bile Duct Neoplasms , Bile Ducts, Intrahepatic , Biomarkers, Tumor , Carcinoma, Hepatocellular , Cholangiocarcinoma , Humans , Liver Neoplasms , Peptides
16.
Strahlenther Onkol ; 192(6): 394-402, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27215563

ABSTRACT

PURPOSE: The benefit of adjuvant chemotherapy (AC) in locoregionally advanced nasopharyngeal carcinoma (NPC) is controversial. This study compared concurrent chemoradiotherapy plus AC (CCRT/AC) with CCRT. METHODS: Pair-matched analysis based on eight clinicopathological features of 244 patients treated with platinum-based CCRT/AC or CCRT alone was performed. Survival outcomes were assessed using the Kaplan-Meier method and log-rank test. Toxicities and response rates were compared using Fisher's exact test. RESULTS: Four-year overall survival, progression-free survival, distant failure-free survival, and locoregional failure-free survival were 72 %, 61 %, 71 %, and 81 %, respectively, for the CCRT arm, compared to 74 % (hazard ratio, HR 0.89; 95 % confidence interval, CI 0.64-1.23; P = 0.474), 62 % (HR 0.91, 95 % CI 0.68-1.20, P = 0.489), 73 % (HR 0.84, 95 % CI 0.59-1.18, P = 0.316), and 84 % (HR 0.84, 95 % CI 0.52-1.24, P = 0.323), respectively, for the CCRT/AC arm. Cox multivariate regression analysis demonstrated AC was not an independent prognostic factor. Overall, there was a higher incidence of grade 3-4 toxicities in the CCRT/AC arm. The most common grade 3-4 adverse events in the CCRT/AC arm were vomiting (27 %), nausea (43 %), leukopenia/neutropenia (23 %), thrombocytopenia (8.8 %), and anemia (6.2 %). CONCLUSION: Addition of AC to CCRT increased toxicities but did not improve survival in locoregionally advanced NPC.


Subject(s)
Chemoradiotherapy/mortality , Chemotherapy, Adjuvant/mortality , Nasopharyngeal Neoplasms/mortality , Nasopharyngeal Neoplasms/therapy , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , China/epidemiology , Female , Humans , Male , Matched-Pair Analysis , Middle Aged , Prevalence , Radiation Injuries/mortality , Risk Factors , Survival Rate , Treatment Outcome , Young Adult
17.
J Exp Med ; 213(5): 859-75, 2016 05 02.
Article in English | MEDLINE | ID: mdl-27091842

ABSTRACT

Oxidative stress status has a key role in hepatocellular carcinoma (HCC) development and progression. Normally, reactive oxygen species (ROS) levels are tightly controlled by an inducible antioxidant program that responds to cellular stressors. How HCC cells respond to excessive oxidative stress remains elusive. Here, we identified a feedback loop between gankyrin, an oncoprotein overexpressed in human HCC, and Nrf2 maintaining the homeostasis in HCC cells. Mechanistically, gankyrin was found to interact with the Kelch domain of Keap1 and effectively competed with Nrf2 for Keap1 binding. Increased expression of gankyrin in HCC cells blocked the binding between Nrf2 and Keap1, inhibiting the degradation of Nrf2 by proteasome. Interestingly, accumulation and translocation of Nrf2 increased the transcription of gankyrin through binding to the ARE elements in the promoter of gankyrin. The positive feedback regulation involving gankyrin and Nrf2 modulates a series of antioxidant enzymes, thereby lowering intracellular ROS and conferring a steadier intracellular environment, which prevents mitochondrial damage and cell death induced by excessive oxidative stress. Our results indicate that gankyrin is a regulator of cellular redox homeostasis and provide a link between oxidative stress and the development of HCC.


Subject(s)
Carcinoma, Hepatocellular/immunology , Liver Neoplasms/immunology , NF-E2-Related Factor 2/immunology , Proteasome Endopeptidase Complex/immunology , Proteolysis , Proto-Oncogene Proteins/immunology , Signal Transduction/immunology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/immunology , Liver Neoplasms/genetics , NF-E2-Related Factor 2/genetics , Oxidative Stress/genetics , Oxidative Stress/immunology , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Proteins/genetics , Reactive Oxygen Species/immunology , Response Elements/immunology , Signal Transduction/genetics
18.
Oncotarget ; 6(5): 3432-42, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25605019

ABSTRACT

Adjuvant transcatheter arterial chemoembolization (TACE) protects against hepatocellular carcinoma (HCC) and is associated with reduced disease recurrence and improved outcome after surgery. However, deterioration of liver function after TACE negatively impacts the patient prognosis and limits it use as an option to prolong survival. We analyzed two independent cohorts that included a total of 510 patients with HCC who had undergone tumor resection. Immunohistochemistry assay was used to measure RPB5-mediating protein (RMP) expression and assessed their association with recurrence rate and response to therapy with adjuvant TACE. In patients with HCC, the expression of RMP in tumor is associated with age, gender, tumor size, portal venous invasion, TNM stages, BCLC stages and overall survival. Among patients with high RMP expression, adjuvant TACE after resection was associated with early recurrence. Even in the patients with small tumor size (no more than 5 cm) or no venous invasion, RMP status is associated with response to adjuvant TACE. RMP status in tumors may be a useful marker in estimating prognosis in patients with HCC and in assisting in the selection of patients who are likely to benefit from adjuvant TACE to prevent relapse.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic , Intracellular Signaling Peptides and Proteins/analysis , Liver Neoplasms/chemistry , Liver Neoplasms/therapy , Adult , Aged , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Hepatectomy , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local , Patient Selection , Predictive Value of Tests , Proportional Hazards Models , Repressor Proteins , Risk Factors , Time Factors , Treatment Outcome , Young Adult
19.
Cancer ; 120(10): 1520-31, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24863391

ABSTRACT

BACKGROUND: The functions of cytoskeleton-associated membrane protein 4 (CKAP4), one kind of type II transmembrane protein, are associated with the palmitoyl acyltransferase DHHC2. The objective of the current study was to investigate CKAP4/DHHC2 expression and its prognostic significance in patients with hepatocellular carcinoma (HCC). METHODS: Two independent cohorts of 416 patients with HCC were enrolled. All the patients included had defined clinicopathologic and follow-up data. Using real-time polymerase chain reaction and immunohistochemical assay, CKAP4 and DHHC2 expression were evaluated. The association between CKAP4/DHHC2 expression and HCC-specific disease-free survival and overall survival was analyzed by Kaplan-Meier curves, the log-rank test, and Multivariate Cox regression analyses. RESULTS: The data documented that CKAP4 expression was much higher in HCC tumor tissues compared with adjacent normal tissues and its expression was significantly correlated with tumor size, intrahepatic metastases, portal venous invasion, and Barcelona Clinic Liver Cancer stage of disease in 2 cohorts of patients. On survival analysis, patients with high CKAP4 expression appeared to have a favorable overall survival and a longer disease-free survival compared with those with low expression. DHHC2 expression was also examined in tissue microarray analysis by immunohistochemistry and the results demonstrated that 87.6% of the cases had low expression of DHHC2. Kaplan-Meier analysis indicated that a high level of DHHC2 expression predicted favorable overall survival and disease-free survival rates in both the training cohort and validation set. Furthermore, the combination of CKAP4 and DHHC2 was found to have a more powerful efficiency in prognosis prediction than either one alone. CONCLUSIONS: To the best of our knowledge, the current study is the first to demonstrate that the expression of CKAP4 and its palmitoyl acyltransferase DHHC2 correlates with disease progression and metastasis in patients with HCC and may provide prognostic and therapeutic value.


Subject(s)
Acyltransferases/analysis , Biomarkers, Tumor/analysis , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/chemistry , Liver Neoplasms/pathology , Membrane Proteins/analysis , Tumor Suppressor Proteins/analysis , Adult , Aged , Biomarkers, Tumor/blood , Blotting, Western , China , Cohort Studies , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Palmitic Acid/metabolism , Portal Vein/pathology , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Tissue Array Analysis
20.
Tumour Biol ; 35(8): 7999-8005, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24838946

ABSTRACT

CKAP4, one kind of type II trans-membrane protein, plays an important role to maintain endoplasmic reticulum structure and inhibits the proliferation of bladder cancer cells by combining its ligand anti-proliferative factor (APF). However, the biological function of CKAP4 in the progression of liver cancer has not been clearly demonstrated. In the present study, we knocked down or overexpressed CKAP4 in hepatocellular carcinoma (HCC) cells and cell proliferation, invasion, and migration capacities were investigated by CCK-8 and transwell assays. In vivo tumor model in mice was used to evaluate the role of CKAP4 on growth and metastasis of HCC. The data documented that HCC cells with high CKAP4 levels were featured by low proliferation capability as well as low invasion potential. Interestingly, we found that CKAP4 suppressed the activation of epithelial growth factor receptor (EGFR) signaling, which may partly explain the role of CKAP4 in cell biological behavior of HCC. Further study revealed that CKAP4 could associate with EGFR at basal status and the complex was reduced upon EGF stimulation, leading to release EGFR into cytoplasm. Thus, we demonstrate the novel mechanism, for the first time, expression of CKAP4 regulates progression and metastasis of HCC and it may provide therapeutic values in this tumor.


Subject(s)
Carcinoma, Hepatocellular/pathology , ErbB Receptors/physiology , Liver Neoplasms/pathology , Membrane Proteins/physiology , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Cell Proliferation , Humans , Male , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL