Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mol Cancer ; 22(1): 147, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37674200

ABSTRACT

Gastric adenocarcinoma (GAC) is a lethal disease characterized by genomic and clinical heterogeneity. By integrating 8 previously established genomic signatures for GAC subtypes, we identified 6 clinically and molecularly distinct genomic consensus subtypes (CGSs). CGS1 have the poorest prognosis, very high stem cell characteristics, and high IGF1 expression, but low genomic alterations. CGS2 is enriched with canonical epithelial gene expression. CGS3 and CGS4 have high copy number alterations and low immune reactivity. However, CGS3 and CGS4 differ in that CGS3 has high HER2 activation, while CGS4 has high SALL4 and KRAS activation. CGS5 has the high mutation burden and moderately high immune reactivity that are characteristic of microsatellite instable tumors. Most CGS6 tumors are positive for Epstein Barr virus and show extremely high levels of methylation and high immune reactivity. In a systematic analysis of genomic and proteomic data, we estimated the potential response rate of each consensus subtype to standard and experimental treatments such as radiation therapy, targeted therapy, and immunotherapy. Interestingly, CGS3 was significantly associated with a benefit from chemoradiation therapy owing to its high basal level of ferroptosis. In addition, we also identified potential therapeutic targets for each consensus subtype. Thus, the consensus subtypes produced a robust classification and provide for additional characterizations for subtype-based customized interventions.


Subject(s)
Adenocarcinoma , Epstein-Barr Virus Infections , Stomach Neoplasms , Humans , Proteomics , Herpesvirus 4, Human , Genomics , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy
2.
Hepatology ; 76(6): 1634-1648, 2022 12.
Article in English | MEDLINE | ID: mdl-35349735

ABSTRACT

BACKGROUND AND AIMS: Although many studies revealed transcriptomic subtypes of HCC, concordance of the subtypes are not fully examined. We aim to examine a consensus of transcriptomic subtypes and correlate them with clinical outcomes. APPROACH AND RESULTS: By integrating 16 previously established genomic signatures for HCC subtypes, we identified five clinically and molecularly distinct consensus subtypes. STM (STeM) is characterized by high stem cell features, vascular invasion, and poor prognosis. CIN (Chromosomal INstability) has moderate stem cell features, but high genomic instability and low immune activity. IMH (IMmune High) is characterized by high immune activity. BCM (Beta-Catenin with high Male predominance) is characterized by prominent ß-catenin activation, low miRNA expression, hypomethylation, and high sensitivity to sorafenib. DLP (Differentiated and Low Proliferation) is differentiated with high hepatocyte nuclear factor 4A activity. We also developed and validated a robust predictor of consensus subtype with 100 genes and demonstrated that five subtypes were well conserved in patient-derived xenograft models and cell lines. By analyzing serum proteomic data from the same patients, we further identified potential serum biomarkers that can stratify patients into subtypes. CONCLUSIONS: Five HCC subtypes are correlated with genomic phenotypes and clinical outcomes and highly conserved in preclinical models, providing a framework for selecting the most appropriate models for preclinical studies.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Male , Female , Carcinoma, Hepatocellular/pathology , beta Catenin/genetics , Liver Neoplasms/pathology , Consensus , Proteomics , Genomics , Phenotype
6.
Cells ; 9(9)2020 09 01.
Article in English | MEDLINE | ID: mdl-32878261

ABSTRACT

AT-rich interactive domain 1A (ARID1A) is one of the most frequently mutated genes in hepatocellular carcinoma (HCC), but its clinical significance is not clarified. We aimed to evaluate the clinical significance of low ARID1A expression in HCC. By analyzing the gene expression data of liver from Arid1a-knockout mice, hepatic Arid1a-specific gene expression signature was identified (p < 0.05 and 0.5-fold difference). From this signature, a prediction model was developed to identify tissues lacking Arid1a activity and was applied to gene expression data from three independent cohorts of HCC patients to stratify patients according to ARID1A activity. The molecular features associated with loss of ARID1A were analyzed using The Cancer Genome Atlas (TCGA) multi-platform data, and Ingenuity Pathway Analysis (IPA) was done to uncover potential signaling pathways associated with ARID1A loss. ARID1A inactivation was clinically associated with poor prognosis in all three independent cohorts and was consistently related to poor prognosis subtypes of previously reported gene signatures (highly proliferative, hepatic stem cell, silence of Hippo pathway, and high recurrence signatures). Immune activity, indicated by significantly lower IFNG6 and cytolytic activity scores and enrichment of regulatory T-cell composition, was lower in the ARID1A-low subtype than ARID1A-high subtype. Ingenuity pathway analysis revealed that direct upstream transcription regulators of the ARID1A signature were genes associated with cell cycle, including E2F group, CCND1, and MYC, while tumor suppressors such as TP53, SMAD3, and CTNNB1 were significantly inhibited. ARID1A plays an important role in immune activity and regulating multiple genes involved in HCC development. Low-ARID1A subtype was associated with poor clinical outcome and suggests the possibility of ARID1A as a prognostic biomarker in HCC patients.


Subject(s)
Carcinoma, Hepatocellular/metabolism , DNA-Binding Proteins/biosynthesis , Liver Neoplasms/metabolism , Transcription Factors/biosynthesis , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genomics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prognosis , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Cells ; 9(8)2020 07 23.
Article in English | MEDLINE | ID: mdl-32717825

ABSTRACT

The critical role of the Hippo pathway has been recently investigated in various cancers, but little is known about its role in glioblastoma (GBM). In order to evaluate the clinical relevance of the Hippo pathway in GBM, we generated a core gene expression signature from four different previously-established silence of Hippo pathway (SOH) signatures. Based on a newly generated core SOH signature, a SOH and active Hippo pathway (AH) was predicted in GBM samples from The Cancer Genome Atlas (TCGA) and validated in a separate cohort. A comparative analysis was performed on multi-panel genomic datasets from TCGA and the possible association of SOH with immune activity and epithelial mesenchymal transition was also evaluated. The SOH signature was associated with poor prognosis in GBM in both cohorts. Expression levels of CTGF and CYR61, the most reliable and well-known downstream targets of YAP1, were markedly increased in the SOH subgroup of GBM patients. SOH signature was strongly associated with a high immune signature score and mesenchymal features. Genes differentially expressed between SOH and AH groups revealed many markers for inhibitory immune checkpoints and M2-polarized macrophages were upregulated in the SOH subgroup, suggesting that SOH may induce the resistance of cancer cells to host immune response in GBM. In summary, SOH is significantly associated with the poor prognosis of GBM patients and is possibly mediated by pro-tumoral immunosuppression.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Immunosuppression Therapy/methods , Protein Serine-Threonine Kinases/genetics , Brain Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Hippo Signaling Pathway , Humans , Male
8.
Biochem Biophys Res Commun ; 511(1): 185-191, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30777332

ABSTRACT

High metabolic activity, reflected in increased glucose uptake, is one of the hallmarks of many cancers including breast cancer. However, not all cancers avidly take up glucose, suggesting heterogeneity in their metabolic demand. Thus, we aim to generate a genomic signature of glucose hypermetabolism in breast cancer and examine its clinical relevance. To identify genes significantly associated with glucose uptake, gene expression data were analyzed together with the standardized uptake values (SUVmax) of 18F-fluorodeoxy-glucose on positron emission tomography (PET) for 11 breast cancers. The resulting PET signature was evaluated for prognostic significance in four large independent patient cohorts (n = 5417). Potential upstream regulators accountable for the high glucose uptake were identified by gene network analysis. A PET signature of 242 genes was significantly correlated with SUVmax in breast cancer. In all four cohorts, high PET signature was significantly associated with poorer prognosis. The prognostic value of this PET signature was further supported by Cox regression analyses (hazard ratio 1.7, confidential interval 1.48-2.02; P < 0.001). The PET signature was also strongly correlated with previously established prognostic genomic signatures such as PAM50, Oncotype DX, and NKI. Gene network analyses suggested that MYC and TBX2 were the most significant upstream transcription factors in the breast cancers with high glucose uptake. A PET signature reflecting high glucose uptake is a novel independent prognostic factor in breast cancer. MYC and TBX2 are potential regulators of glucose uptake.


Subject(s)
Breast Neoplasms/metabolism , Glucose/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Female , Fluorodeoxyglucose F18/metabolism , Gene Expression Regulation, Neoplastic , Glucose/genetics , Glycolysis , Humans , Positron-Emission Tomography/methods , Prognosis
9.
Oral Oncol ; 85: 44-51, 2018 10.
Article in English | MEDLINE | ID: mdl-30220319

ABSTRACT

OBJECTIVES: Heterogeneity of head and neck squamous cell carcinomas (HNSCCs) results in unpredictable outcomes for patients with similar stages of cancer. Beyond the role of human papilloma virus (HPV), no validated molecular marker of HNSCCs has been established. Thus, clinically relevant molecular subtypes are needed to optimize HNSCC therapy. The purpose of this study was to identify subtypes of HNSCC that have distinct biological characteristics associated with clinical outcomes and to characterize genomic alterations that best reflect the biological and clinical characteristics of each subtype. MATERIALS AND METHODS: We analyzed gene expression profiling data from pan-SCC tissues including cervical SCC, esophageal SCC, lung SCC, and HNSCC (n = 1346) to assess the similarities and differences among SCCs and to identify molecular subtypes of HNSCC associated with prognosis. Subtype-specific gene expression signatures were identified and used to construct predictive models. The association of the subtypes with prognosis was validated in two independent cohorts of patients. RESULTS: Pan-SCC analysis identified three novel subtypes of HNSCC. Subtype 1 had the best prognosis and was similar to cervical SCC, whereas subtype 3 had the worst prognosis and was similar to lung SCC. Subtype 2 had a moderate prognosis. The 600-gene signature associated with the three subtypes significantly predicted prognosis in two independent validation cohorts. These three subtypes also were associated with potential benefit of immunotherapy. CONCLUSION: We identified three clinically relevant HNSCC molecular subtypes. Independent prospective studies to assess the clinical utility of the subtypes and associated gene signature are warranted.


Subject(s)
Carcinoma, Squamous Cell/classification , Head and Neck Neoplasms/classification , Squamous Cell Carcinoma of Head and Neck/classification , Aged , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/therapy , Cluster Analysis , Cohort Studies , Female , Gene Dosage , Gene Expression Profiling , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/therapy , Humans , Immunotherapy , Kaplan-Meier Estimate , Male , Middle Aged , Papillomavirus Infections/genetics , Papillomavirus Infections/mortality , Prognosis , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/therapy , Treatment Outcome
10.
Cancer Res ; 78(12): 3135-3146, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29724720

ABSTRACT

Loss of the short arm of chromosome 3 (3p) occurs early in >95% of clear cell renal cell carcinoma (ccRCC). Nearly ubiquitous 3p loss in ccRCC suggests haploinsufficiency for 3p tumor suppressors as early drivers of tumorigenesis. We previously reported methyltransferase SETD2, which trimethylates H3 histones on lysine 36 (H3K36me3) and is located in the 3p deletion, to also trimethylate microtubules on lysine 40 (αTubK40me3) during mitosis, with αTubK40me3 required for genomic stability. We now show that monoallelic, Setd2-deficient cells retaining H3K36me3, but not αTubK40me3, exhibit a dramatic increase in mitotic defects and micronuclei count, with increased viability compared with biallelic loss. In SETD2-inactivated human kidney cells, rescue with a pathogenic SETD2 mutant deficient for microtubule (αTubK40me3), but not histone (H3K36me3) methylation, replicated this phenotype. Genomic instability (micronuclei) was also a hallmark of patient-derived cells from ccRCC. These data show that the SETD2 tumor suppressor displays a haploinsufficiency phenotype disproportionately impacting microtubule methylation and serves as an early driver of genomic instability.Significance: Loss of a single allele of a chromatin modifier plays a role in promoting oncogenesis, underscoring the growing relevance of tumor suppressor haploinsufficiency in tumorigenesis. Cancer Res; 78(12); 3135-46. ©2018 AACR.


Subject(s)
Carcinoma, Renal Cell/genetics , Chromosomes, Human, Pair 3/genetics , Histone-Lysine N-Methyltransferase/genetics , Kidney Neoplasms/genetics , Microtubules/metabolism , Animals , Carcinogenesis/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Fibroblasts , Gene Knockdown Techniques , Genomic Instability , Haploinsufficiency , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Kidney Neoplasms/pathology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/pathology , Lysine/metabolism , Methylation , Mice , Micronuclei, Chromosome-Defective
11.
Nat Commun ; 9(1): 1777, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29725014

ABSTRACT

Gastric cancer is a heterogeneous cancer, making treatment responses difficult to predict. Here we show that we identify two distinct molecular subtypes, mesenchymal phenotype (MP) and epithelial phenotype (EP), by analyzing genomic and proteomic data. Molecularly, MP subtype tumors show high genomic integrity characterized by low mutation rates and microsatellite stability, whereas EP subtype tumors show low genomic integrity. Clinically, the MP subtype is associated with markedly poor survival and resistance to standard chemotherapy, whereas the EP subtype is associated with better survival rates and sensitivity to chemotherapy. Integrative analysis shows that signaling pathways driving epithelial-to-mesenchymal transition and insulin-like growth factor 1 (IGF1)/IGF1 receptor (IGF1R) pathway are highly activated in MP subtype tumors. Importantly, MP subtype cancer cells are more sensitive to inhibition of IGF1/IGF1R pathway than EP subtype. Detailed characterization of these two subtypes could identify novel therapeutic targets and useful biomarkers for prognosis and therapy response.


Subject(s)
Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Gene Expression Regulation, Neoplastic , Mesoderm/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Chemotherapy, Adjuvant , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Female , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/metabolism , Heterografts , Humans , Kaplan-Meier Estimate , Mice, Inbred BALB C , Microsatellite Instability , Mutation , Prognosis , Proteomics , Receptor, IGF Type 1/metabolism , Reproducibility of Results , Signal Transduction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism
12.
Exp Mol Med ; 50(1): e421, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29303508

ABSTRACT

The gene encoding ß-catenin is frequently mutated in hepatocellular carcinoma cells. While the oncogenicity of ß-catenin has been extensively studied, ß-catenin's role in hepatocellular carcinoma tumor metabolism is currently less well understood. In this study, we found that ß-catenin regulates the expression of glutamine synthetase and triggers a series of metabolic changes leading to induction of autophagy in hepatocellular carcinoma cells. ß-Catenin-active Hep3B and HepG2 cells exhibited higher basal levels of autophagic activity than did ß-catenin wild-type cells. We also found that autophagy in ß-catenin-active cells was mediated by glutamine synthetase, as silencing of glutamine synthetase significantly reduced autophagic activity. We also showed that ß-catenin-active hepatocellular carcinoma cells were more sensitive to sorafenib than were ß-catenin wild-type cells. Our results demonstrated that glutamine synthetase-mediated autophagy explains the high sensitivity of ß-catenin-active hepatocellular carcinoma cells to sorafenib. Our results highlight the importance of glutamine metabolism in the regulation of autophagy in hepatocellular carcinoma cells. More importantly, our study unravels the molecular mechanisms leading to sorafenib sensitivity in hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Glutamate-Ammonia Ligase/metabolism , Liver Neoplasms/drug therapy , Sorafenib/pharmacology , beta Catenin/metabolism , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy/physiology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Glutamate-Ammonia Ligase/genetics , Humans , Liver Neoplasms/metabolism
13.
Nat Commun ; 8(1): 1050, 2017 10 19.
Article in English | MEDLINE | ID: mdl-29051489

ABSTRACT

Immunotherapy has emerged as a promising anti-cancer treatment, however, little is known about the genetic characteristics that dictate response to immunotherapy. We develop a transcriptional predictor of immunotherapy response and assess its prediction in genomic data from ~10,000 human tissues across 30 different cancer types to estimate the potential response to immunotherapy. The integrative analysis reveals two distinct tumor types: the mutator type is positively associated with potential response to immunotherapy, whereas the chromosome-instable type is negatively associated with it. We identify somatic mutations and copy number alterations significantly associated with potential response to immunotherapy, in particular treatment with anti-CTLA-4 antibody. Our findings suggest that tumors may evolve through two different paths that would lead to marked differences in immunotherapy response as well as different strategies for evading immune surveillance. Our analysis provides resources to facilitate the discovery of predictive biomarkers for immunotherapy that could be tested in clinical trials.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Gene Dosage , Neoplasms/therapy , Animals , Antibodies/therapeutic use , CTLA-4 Antigen/immunology , Gene Expression , Genetic Markers , Genomics , Humans , Immunotherapy , Mice , Mutation , Neoplasms/classification , Neoplasms/genetics , Neoplasms/virology , Treatment Outcome
14.
Clin Cancer Res ; 23(15): 4441-4449, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28747339

ABSTRACT

Purpose: The Cancer Genome Atlas (TCGA) project recently uncovered four molecular subtypes of gastric cancer: Epstein-Barr virus (EBV), microsatellite instability (MSI), genomically stable (GS), and chromosomal instability (CIN). However, their clinical significances are currently unknown. We aimed to investigate the relationship between subtypes and prognosis of patients with gastric cancer.Experimental Design: Gene expression data from a TCGA cohort (n = 262) were used to develop a subtype prediction model, and the association of each subtype with survival and benefit from adjuvant chemotherapy was tested in 2 other cohorts (n = 267 and 432). An integrated risk assessment model (TCGA risk score) was also developed.Results: EBV subtype was associated with the best prognosis, and GS subtype was associated with the worst prognosis. Patients with MSI and CIN subtypes had poorer overall survival than those with EBV subtype but better overall survival than those with GS subtype (P = 0.004 and 0.03 in two cohorts, respectively). In multivariate Cox regression analyses, TCGA risk score was an independent prognostic factor [HR, 1.5; 95% confidence interval (CI), 1.2-1.9; P = 0.001]. Patients with the CIN subtype experienced the greatest benefit from adjuvant chemotherapy (HR, 0.39; 95% CI, 0.16-0.94; P = 0.03) and those with the GS subtype had the least benefit from adjuvant chemotherapy (HR, 0.83; 95% CI, 0.36-1.89; P = 0.65).Conclusions: Our prediction model successfully stratified patients by survival and adjuvant chemotherapy outcomes. Further development of the prediction model is warranted. Clin Cancer Res; 23(15); 4441-9. ©2017 AACR.

15.
Oncotarget ; 8(67): 111130-111143, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29340043

ABSTRACT

By analyzing the genomic data of head and neck squamous cell cancer (HNSCC), we investigated clinical significance of YAP1 activation. Copy number and mRNA expression of YAP1 were analyzed together to assess clinical relevance of YAP1 activation in HNSCC. The clinical significance of YAP1 activation was further validated in four independent test cohorts. We also assessed the correlation of YAP1 activation with genomic alterations such as copy number alteration, somatic mutation, and miRNA expression. The YAP1-activated (YA) subgroup showed worse prognosis for HNSCC as tested and validated in five cohorts. In a multivariate risk analysis, the YAP1 signature was the most significant predictor of overall survival. The YAP1-inactivated (YI) subgroup was associated with HPV-positive status. In multiplatform analysis, YA tumors had gain of EGFR and SNAI2; loss of tumor-suppressor genes such as CSMD1, CDKN2A, NOTCH1, and SMAD4; and high mutation rates of TP53 and CDKN2A. YI tumors were characterized by gain of PIK3CA, SOX2, and TP63; deletion of 11q23.1; and high mutation rates of NFE2L2, PTEN, SYNE1, and NSD1. YA tumors also showed weaker immune activity as reflected in low IFNG composite scores and YAP1 activity is negatively associated with potential response to treatment of pembrolizumab. In conclusion, activation of YAP1 is associated with worse prognosis of patients with HNSCC and potential resistance to immunotherapy.

16.
Gut ; 66(2): 215-225, 2017 02.
Article in English | MEDLINE | ID: mdl-27507904

ABSTRACT

OBJECTIVE: Oesophageal squamous cell carcinoma (ESCC) is a heterogeneous disease with variable outcomes that are challenging to predict. A better understanding of the biology of ESCC recurrence is needed to improve patient care. Our goal was to identify small non-coding RNAs (sncRNAs) that could predict the likelihood of recurrence after surgical resection and to uncover potential molecular mechanisms that dictate clinical heterogeneity. DESIGN: We developed a robust prediction model for recurrence based on the analysis of the expression profile data of sncRNAs from 108 fresh frozen ESCC specimens as a discovery set and assessment of the associations between sncRNAs and recurrence-free survival (RFS). We also evaluated the mechanistic and therapeutic implications of sncRNA obtained through integrated analysis from multiple datasets. RESULTS: We developed a risk assessment score (RAS) for recurrence with three sncRNAs (microRNA (miR)-223, miR-1269a and nc886) whose expression was significantly associated with RFS in the discovery cohort (n=108). RAS was validated in an independent cohort of 512 patients. In multivariable analysis, RAS was an independent predictor of recurrence (HR, 2.27; 95% CI, 1.26 to 4.09; p=0.007). This signature implies the expression of ΔNp63 and multiple alterations of driver genes like PIK3CA. We suggested therapeutic potentials of immune checkpoint inhibitors in low-risk patients, and Polo-like kinase inhibitors, mammalian target of rapamycin (mTOR) inhibitors, and histone deacetylase inhibitors in high-risk patients. CONCLUSION: We developed an easy-to-use prognostic model with three sncRNAs as robust prognostic markers for postoperative recurrence of ESCC. We anticipate that such a stratified and systematic, tumour-specific biological approach will potentially contribute to significant improvement in ESCC treatment.


Subject(s)
Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , MicroRNAs/analysis , Neoplasm Recurrence, Local/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/surgery , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor/drug effects , Class I Phosphatidylinositol 3-Kinases , Disease-Free Survival , Drug Screening Assays, Antitumor , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/surgery , Female , Genomics , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , MicroRNAs/genetics , Middle Aged , Models, Biological , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases/genetics , Predictive Value of Tests , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Risk Assessment , Systems Biology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Polo-Like Kinase 1
17.
Clin Cancer Res ; 22(24): 6228-6235, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27654712

ABSTRACT

PURPOSE: This study was aimed at developing and validating a quantitative multigene assay for predicting tumor recurrence after gastric cancer surgery. EXPERIMENTAL DESIGN: Gene expression data were generated from tumor tissues of patients who underwent surgery for gastric cancer (n = 267, training cohort). Genes whose expression was significantly associated with activation of YAP1 (a frequently activated oncogene in gastrointestinal cancer), 5-year recurrence-free survival, and 5-year overall survival were first identified as candidates for prognostic genes (156 genes, P < 0.001). We developed the recurrence risk score (RRS) by using quantitative RT-PCR to identify genes whose expression levels were significantly associated with YAP1 activation and patient survival in the training cohort. RESULTS: We based the RRS assay on 6 genes, IGFBP4, SFRP4, SPOCK1, SULF1, THBS, and GADD45B, whose expression levels were significantly associated with YAP1 activation and prognosis in the training cohort. The RRS assay was further validated in an independent cohort of 317 patients. In multivariate analysis, the RRS was an independent predictor of recurrence [HR, 1.6; 95% confidence interval (CI), 1.02-2.4; P = 0.03]. In patients with stage II disease, the RRS had an HR of 2.9 (95% CI, 1.1-7.9; P = 0.03) and was the only significant independent predictor of recurrence. CONCLUSIONS: The RRS assay was a valid predictor of recurrence in the two cohorts of patients with gastric cancer. Independent prospective studies to assess the clinical utility of this assay are warranted. Clin Cancer Res; 22(24); 6228-35. ©2016 AACR.


Subject(s)
Neoplasm Recurrence, Local/genetics , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Gene Expression/genetics , Gene Expression Profiling/methods , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Prognosis , Risk , Stomach Neoplasms/pathology
18.
Hepatology ; 64(5): 1757-1772, 2016 11.
Article in English | MEDLINE | ID: mdl-27531557

ABSTRACT

In the adult liver, the Hippo pathway mammalian STE20-like protein kinases 1 and 2 and large tumor suppressor homologs 1 and 2 (LATS1/2) control activation of the transcriptional coactivators Yes-associated protein (YAP) and WW domain containing transcription regulator 1 (TAZ) in hepatocytes and biliary epithelial cells, thereby regulating liver cell proliferation, differentiation, and malignant transformation. Less is known about the contribution of Hippo signaling to liver development. We used conditional mutagenesis to show that the Hippo signaling pathway kinases LATS1 and LATS2 are redundantly required during mouse liver development to repress YAP and TAZ in both the biliary epithelial and hepatocyte lineages. In the absence of LATS1/2, biliary epithelial cells exhibit excess proliferation while hepatoblasts fail to mature into hepatocytes, defects that result in perinatal lethality. Using an in vitro hepatocyte differentiation assay, we demonstrate that YAP activity decreases and Hippo pathway kinase activities increase upon differentiation. In addition, we show that YAP activation in vitro, resulting from either depletion of its negative regulators LATS1/2 or expression of a mutant form of YAP that is less efficiently phosphorylated by LATS1/2, results in transcriptional suppression of genes that normally accompany hepatocyte maturation. Moreover, we provide evidence that YAP activity is repressed by Hippo pathway activation upon hepatocytic maturation in vitro. Finally, we examine the localization of YAP during fetal liver development and show that higher levels of YAP are found in biliary epithelial cells, while in hepatocytes YAP levels decrease with hepatocyte maturation. CONCLUSION: Hippo signaling, mediated by the LATS1 and LATS2 kinases, is required to restrict YAP and TAZ activation during both biliary and hepatocyte differentiation. These findings suggest that dynamic regulation of the Hippo signaling pathway plays an important role in differentiation and functional maturation of the liver. (Hepatology 2016;64:1757-1772).


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Proliferation/physiology , Hepatocytes/physiology , Liver/cytology , Phosphoproteins/physiology , Protein Serine-Threonine Kinases/physiology , Stem Cells/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Cycle Proteins , Cell Differentiation , Mice , Trans-Activators , YAP-Signaling Proteins
19.
Clin Cancer Res ; 22(5): 1256-64, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26459179

ABSTRACT

PURPOSE: The Hippo pathway is a tumor suppressor in the liver. However, the clinical significance of Hippo pathway inactivation in HCC is not clearly defined. We analyzed genomic data from human and mouse tissues to determine clinical relevance of Hippo pathway inactivation in HCC. EXPERIMENTAL DESIGN: We analyzed gene expression data from Mst1/2(-/-) and Sav1(-/-) mice and identified a 610-gene expression signature reflecting Hippo pathway inactivation in the liver [silence of Hippo (SOH) signature]. By integrating gene expression data from mouse models with those from human HCC tissues, we developed a prediction model that could identify HCC patients with an inactivated Hippo pathway and used it to test its significance in HCC patients, via univariate and multivariate Cox analyses. RESULTS: HCC patients (National Cancer Institute cohort, n = 113) with the SOH signature had a significantly poorer prognosis than those without the SOH signature [P < 0.001 for overall survival (OS)]. The significant association of the signature with poor prognosis was further validated in the Korean (n = 100, P = 0.006 for OS) and Fudan University cohorts (n = 242, P = 0.001 for OS). On multivariate analysis, the signature was an independent predictor of recurrence-free survival (HR, 1.6; 95% confidence interval, 1.12-2.28: P = 0.008). We also demonstrated significant concordance between the SOH HCC subtype and the hepatic stem cell HCC subtype that had been identified in a previous study (P < 0.001). CONCLUSIONS: Inactivation of the Hippo pathway in HCC is significantly associated with poor prognosis.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Phosphoproteins/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Kaplan-Meier Estimate , Liver Neoplasms/pathology , Male , Mice , Middle Aged , Phosphoproteins/genetics , Prognosis , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
20.
Hepatology ; 63(1): 159-72, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26389641

ABSTRACT

UNLABELLED: Metabolic activation is a common feature of many cancer cells and is frequently associated with the clinical outcomes of various cancers, including hepatocellular carcinoma. Thus, aberrantly activated metabolic pathways in cancer cells are attractive targets for cancer therapy. Yes-associated protein 1 (YAP1) and transcriptional coactivator with PDZ-binding motif (TAZ) are oncogenic downstream effectors of the Hippo tumor suppressor pathway, which is frequently inactivated in many cancers. Our study revealed that YAP1/TAZ regulates amino acid metabolism by up-regulating expression of the amino acid transporters solute carrier family 38 member 1 (SLC38A1) and solute carrier family 7 member 5 (SLC7A5). Subsequently, increased uptake of amino acids by the transporters (SLC38A1 and SLC7A5) activates mammalian target of rapamycin complex 1 (mTORC1), a master regulator of cell growth, and stimulates cell proliferation. We also show that high expression of SLC38A1 and SLC7A5 is significantly associated with shorter survival in hepatocellular carcinoma patients. Furthermore, inhibition of the transporters and mTORC1 significantly blocks YAP1/TAZ-mediated tumorigenesis in the liver. These findings elucidate regulatory networks connecting the Hippo pathway to mTORC1 through amino acid metabolism and the mechanism's potential clinical implications for treating hepatocellular carcinoma. CONCLUSION: YAP1 and TAZ regulate cancer metabolism and mTORC1 through regulation of amino acid transportation, and two amino acid transporters, SLC38A1 and SLC7A5, might be important therapeutic targets.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Amino Acid Transport Systems/physiology , Carcinoma, Hepatocellular/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Liver Neoplasms/metabolism , Multiprotein Complexes/physiology , Phosphoproteins/physiology , TOR Serine-Threonine Kinases/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinoma, Hepatocellular/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Mechanistic Target of Rapamycin Complex 1 , Mice , Phosphoproteins/genetics , Protein Structure, Tertiary , Signal Transduction , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...