Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
J Affect Disord Rep ; 12: 100494, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36777966

ABSTRACT

Background: India has been badly affected by Covid-19 not only in terms of human lives but also has a long-term effect on mental health of the population. This paper is an attempt to understand the psychological effects of the pandemic on the college and university students in India after the second wave of COVID-19 outbreak and its associated factors. Method: A web-based survey was conducted to collect information from the students both at individual and household level. The study applied binary and multivariate logistic regression to estimate the adjusted and unadjusted marginal effects of the predictor variables. Result: Results show a significant increase in mental health concerns during the second wave of the pandemic, as compared to the first year. The key factors contributing to the higher prevalence of depression, anxiety, and stress are the difficulties faced in the adaptation to the new way of learning, fear of discontinuation of education due to financial constraints faced by household, limited physical interaction, and prolonged screen-time during the pandemic. Limitation: The study has some limitations regarding selection of the sample as the survey was web-based. Also, the mental health situation of the students is self-reported and the study does not consider the prevailing mental health issues before the pandemic. Conclusion: The study recommends initiatives like offering counselling classes and strategies to cope up with mental health disorders. Further, there is a need to conduct follow-up studies to assess the long-term impacts of prolonged home quarantine on the mental health of the students.

2.
J Clin Invest ; 130(6): 3005-3020, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32364535

ABSTRACT

Transcriptional reactivation of telomerase catalytic subunit (TERT) is a frequent hallmark of cancer, occurring in 90% of human malignancies. However, specific mechanisms driving TERT reactivation remain obscure for many tumor types and in particular gastric cancer (GC), a leading cause of global cancer mortality. Here, through comprehensive genomic and epigenomic analysis of primary GCs and GC cell lines, we identified the transcription factor early B cell factor 1 (EBF1) as a TERT transcriptional repressor and inactivation of EBF1 function as a major cause of TERT upregulation. Abolishment of EBF1 function occurs through 3 distinct (epi)genomic mechanisms. First, EBF1 is epigenetically silenced via DNA methyltransferase, polycomb-repressive complex 2 (PRC2), and histone deacetylase activity in GCs. Second, recurrent, somatic, and heterozygous EBF1 DNA-binding domain mutations result in the production of dominant-negative EBF1 isoforms. Third, more rarely, genomic deletions and rearrangements proximal to the TERT promoter remobilize or abolish EBF1-binding sites, derepressing TERT and leading to high TERT expression. EBF1 is also functionally required for various malignant phenotypes in vitro and in vivo, highlighting its importance for GC development. These results indicate that multimodal genomic and epigenomic alterations underpin TERT reactivation in GC, converging on transcriptional repressors such as EBF1.


Subject(s)
Epigenomics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Stomach Neoplasms/metabolism , Telomerase/biosynthesis , Trans-Activators/metabolism , Cell Line, Tumor , Humans , Mutation , Neoplasm Proteins/genetics , Response Elements , Stomach Neoplasms/genetics , Telomerase/genetics , Trans-Activators/genetics
3.
Mod Pathol ; 33(10): 2075-2086, 2020 10.
Article in English | MEDLINE | ID: mdl-32269290

ABSTRACT

Spasmolytic polypeptide-expressing metaplasia (SPEM) and pyloric gland adenoma (PGA) in the stomach are metaplastic and neoplastic lesions, respectively, in which gastric body glands are replaced by pyloric glands. The aim of this study was to evaluate the genomic profile of SPEM and compare it with intestinal-type gastric cancer (GC) and PGA. Thirteen gastrectomies showing PGA with or without dysplasia, GC and SPEM were retrospectively selected. MUC5AC, MUC6, gastrin, and TFF2 IHC were performed. Lesions were subjected to laser capture microdissection followed by DNA extraction. Forty-three DNA samples were extracted from PGA without cytological dysplasia, PGA with low-grade and high-grade dysplasia and pyloric gland adenocarcinoma, GC, SPEM, and adjacent normal tissue from the body of the stomach and were subjected to exome sequencing for 49 genes that are commonly dysregulated in GC. Sanger sequencing was performed for confirmation. Twenty nonsynonymous mutations were identified in SPEM, and none of these were frameshifts or indels. PGA with or without cytological dysplasia showed a significantly higher number of mutations compared with SPEM. As cytological dysplasia increased from no dysplasia to dysplasia in PGA, the percentage of frameshift mutations, indels, and missense variations increased. Further missense or frameshift mutations were observed in the KRAS, APC, TP53, and CTNNB1 genes in the PGA group. In GC, mutations were observed in the TP53 gene (p.Arg248Gln). Missense mutations in the MUC5AC, KRAS, BRAF, and EZH2 genes were common between SPEM and GC. SPEM showed fewer genomic variations than GC and PGA, and was genomically distinct from the pyloric epithelium in PGA. Stepwise progression of PGA from PGA without dysplasia to PGA with dysplasia/adenocarcinoma was associated an increase in mutations. SPEM appears to be more genomically similar to GC than PGA.


Subject(s)
Adenoma/genetics , Gastric Mucosa/pathology , Precancerous Conditions/genetics , Stomach Diseases/genetics , Stomach Neoplasms/genetics , Adenoma/pathology , Humans , Laser Capture Microdissection , Metaplasia/genetics , Metaplasia/pathology , Mutation , Precancerous Conditions/pathology , Retrospective Studies , Singapore , Stomach/pathology , Stomach Diseases/pathology , Stomach Neoplasms/pathology
4.
NAR Genom Bioinform ; 2(2): lqaa013, 2020 Jun.
Article in English | MEDLINE | ID: mdl-33575575

ABSTRACT

Comprehensive understanding of aberrant splicing in gastric cancer is lacking. We RNA-sequenced 19 gastric tumor-normal pairs and identified 118 high-confidence tumor-associated (TA) alternative splicing events (ASEs) based on high-coverage sequencing and stringent filtering, and also identified 8 differentially expressed splicing factors (SFs). The TA ASEs occurred in genes primarily involved in cytoskeletal organization. We constructed a correlative network between TA ASE splicing ratios and SF expression, replicated it in independent gastric cancer data from The Cancer Genome Atlas and experimentally validated it by knockdown of the nodal SFs (PTBP1, ESRP2 and MBNL1). Each SF knockdown drove splicing alterations in several corresponding TA ASEs and led to alterations in cellular migration consistent with the role of TA ASEs in cytoskeletal organization. We have therefore established a robust network of dysregulated splicing associated with tumor invasion in gastric cancer. Our work is a resource for identifying oncogenic splice forms, SFs and splicing-generated tumor antigens as biomarkers and therapeutic targets.

5.
Gut ; 69(2): 231-242, 2020 02.
Article in English | MEDLINE | ID: mdl-31068366

ABSTRACT

OBJECTIVE: Gastric cancer (GC) is a leading cause of cancer mortality. Previous studies have shown that hepatocyte nuclear factor-4α (HNF4α) is specifically overexpressed in GC and functionally required for GC development. In this study, we investigated, on a genome-wide scale, target genes of HNF4α and oncogenic pathways driven by HNF4α and HNF4α target genes. DESIGN: We performed HNF4α chromatin immunoprecipitation followed by sequencing across multiple GC cell lines, integrating HNF4α occupancy data with (epi)genomic and transcriptome data of primary GCs to define HNF4α target genes of in vitro and in vivo relevance. To investigate mechanistic roles of HNF4α and HNF4α targets, we performed cancer metabolic measurements, drug treatments and functional assays including murine xenograft experiments. RESULTS: Gene expression analysis across 19 tumour types revealed HNF4α to be specifically upregulated in GCs. Unbiased pathway analysis revealed organic acid metabolism as the top HNF4α-regulated pathway, orthogonally supported by metabolomic analysis. Isocitrate dehydrogenase 1 (IDH1) emerged as a convergent HNF4α direct target gene regulating GC metabolism. We show that wild-type IDH1 is essential for GC cell survival, and that certain GC cells can be targeted by IDH1 inhibitors. CONCLUSIONS: Our results highlight a role for HNF4α in sustaining GC oncogenic metabolism, through the regulation of IDH1. Drugs targeting wild-type IDH1 may thus have clinical utility in GCs exhibiting HNF4α overexpression, expanding the role of IDH1 in cancer beyond IDH1/2 mutated malignancies.


Subject(s)
Hepatocyte Nuclear Factor 4/genetics , Isocitrate Dehydrogenase/metabolism , Stomach Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Targeting/methods , Hepatocyte Nuclear Factor 4/metabolism , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Mice, Inbred NOD , Molecular Targeted Therapy/methods , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Promoter Regions, Genetic/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Up-Regulation/genetics , Xenograft Model Antitumor Assays
6.
Pathol Oncol Res ; 25(1): 333-340, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29116623

ABSTRACT

Microsatellite instability (MSI) is one of the subgroups based on the new molecular classification of gastric cancer (GC). In this study, we analyzed the role of KRAS status in MSI GC and the impact of MSI status on KRAS mutation. We performed analysis on 595 GC patients. Polymerase chain reaction (PCR) was used for the screening of KRAS mutation (exon 2) and 5 quasi-monomorphic mononucleotide repeats, namely, BAT-26, BAT-25, NR -24, NR-21, and NR-27 were used to determine the MSI status. The KRAS and MSI status were then compared with clinicopathologic data of the GC patients. MSI GC was found in 20.3% of all cases. KRAS mutation was seen in 24 patients; 18 were MSI (75%) and 6 were microsatellite stable (MSS) (25%). MSI GC patients with KRAS mutation were older and mostly female, but MSS presented more advanced T and N stage of the disease, more cardia tumors, and adjuvant treatment. Five-year survival was 72.2% for KRAS mutation patients with MSI and 0% for MSS (p < 0.001). Although KRAS mutations in GC are linked with MSI in the majority of cases, KRAS mutations with MSS status presented with a poor prognosis and a worse outcome. In multivariate analysis, MSI was associated with better survival (p < 0.001) but KRAS was with worse survival (p = 0.304). Our study suggests that KRAS mutations are based on MSI status rather than different codon subtypes of mutation, and such a division could be used to determine the GC patient outcome.


Subject(s)
Biomarkers, Tumor/genetics , Microsatellite Instability , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Stomach Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Adenocarcinoma, Mucinous/surgery , Aged , Aged, 80 and over , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , Carcinoma, Signet Ring Cell/surgery , Female , Follow-Up Studies , Humans , Male , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/surgery , Survival Rate
7.
Gastric Cancer ; 22(1): 77-90, 2019 01.
Article in English | MEDLINE | ID: mdl-29779068

ABSTRACT

BACKGROUND: Gastric cancer with lymphoid stroma (GCLS) is characterized by prominent stromal infiltration of T-lymphocytes. The aim of this study was to investigate GCLS biology through analysis of clinicopathological features, EBV infection, microsatellite instability (MSI), immune gene-expression profiling and PD-L1 status in neoplastic cells and tumor immune microenvironment. METHODS: Twenty-four GCLSs were analyzed by RNA in situ hybridization for EBV (EBER), PCR/fragment analysis for MSI, immunohistochemistry (PD-L1, cytokeratin, CD3, CD8), co-immunofluorescence (CK/PD-L1, CD68/PD-L1), NanoString gene-expression assay for immune-related genes and PD-L1 copy number alterations. CD3+ and CD8+ T-cell densities were calculated by digital analysis. Fifty-four non-GCLSs were used as control group. RESULTS: GCLSs displayed distinctive clinicopathological features, such as lower pTNM stage (p = 0.02) and better overall survival (p = 0.01). EBV+ or MSI-high phenotype was found in 66.7 and 16.7% cases, respectively. GCLSs harbored a cytotoxic T-cell-inflamed profile, particularly at the invasive front of tumors (p < 0.01) and in EBV+ cases (p = 0.01). EBV+ GCLSs, when compared to EBV- GCLSs, showed higher mRNA expression of genes related to Th1/cytotoxic and immunosuppressive biomarkers. PD-L1 protein expression, observed in neoplastic and immune stromal cells (33.3 and 91.7%, respectively), and PD-L1 amplification (18.8%) were restricted to EBV+/MSI-high tumors and correlated with high values of PD-L1 mRNA expression. CONCLUSIONS: This study shows that GCLS has a distinctive clinico-pathological and molecular profile. Furthermore, through an in-depth study of tumor immune microenvironment-by digital analysis and mRNA expression profiling-it highlights the role of EBV infection in promoting an inflamed tumor microenvironment, with putative therapeutic implications.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Stomach Neoplasms/pathology , Tumor Microenvironment/immunology , Adult , Aged , B7-H1 Antigen/biosynthesis , Epstein-Barr Virus Infections/complications , Female , Herpesvirus 4, Human , Humans , Immunophenotyping , Inflammation/genetics , Inflammation/immunology , Male , Microsatellite Instability , Middle Aged , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transcriptome , Tumor Microenvironment/genetics
8.
Int J Mol Sci ; 19(7)2018 Jul 17.
Article in English | MEDLINE | ID: mdl-30018250

ABSTRACT

BACKGROUND: Epstein-Barr Virus (EBV) positive and microsatellite unstable (MSI-high) gastric cancer (GC) are molecular subgroups with distinctive molecular profiles. We explored the transcriptomic differences between EBV+ and MSI-high GCs, and the expression of current GC immunotherapy targets such as PD-1, PD-L1, CTLA4 and Dies1/VISTA. METHODS: Using Nanostring Technology and comparative bioinformatics, we analyzed the expression of 499 genes in 46 GCs, classified either as EBV positive (EBER in situ hybridization) or MSI-high (PCR/fragment analysis). PD-L1 protein expression was assessed by immunohistochemistry. RESULTS: From the 46 GCs, 27 tested MSI-high/EBV-, 15 tested MSS/EBV+ and four tested MSS/EBV-. The Nanostring CodeSet could segregate GCs according to MSI and, to a lesser extent, EBV status. Functional annotation of differentially expressed genes associated MSI-high/EBV- GCs with mitotic activity and MSS/EBV+ GCs with immune response. PD-L1 protein expression, evaluated in stromal immune cells, was lower in MSI-high/EBV- GCs. High mRNA expression of PD-1, CTLA4 and Dies1/VISTA and distinctive PD-1/PD-L1 co-expression patterns (PD-1high/PD-L1low, PD-1high/PDL1high) were associated with MSS/EBV+ molecular subtype and gastric cancer with lymphoid stroma (GCLS) morphological features. CONCLUSIONS: EBV+ and MSI-high GCs present distinct transcriptomic profiles. GCLS/EBV+ cases frequently present co-expression of multiple immunotherapy targets, a finding with putative therapeutic implications.


Subject(s)
Epstein-Barr Virus Infections/genetics , Gene Expression Regulation, Neoplastic , Microsatellite Instability , Stomach Neoplasms/genetics , Transcriptome , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cluster Analysis , Epstein-Barr Virus Infections/virology , Gene Expression Profiling/methods , Gene Ontology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Retrospective Studies , Stomach Neoplasms/virology
9.
Cancer Cell ; 33(1): 137-150.e5, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29290541

ABSTRACT

Intestinal metaplasia (IM) is a pre-malignant condition of the gastric mucosa associated with increased gastric cancer (GC) risk. We performed (epi)genomic profiling of 138 IMs from 148 cancer-free patients, recruited through a 10-year prospective study. Compared with GCs, IMs exhibit low mutational burdens, recurrent mutations in certain tumor suppressors (FBXW7) but not others (TP53, ARID1A), chromosome 8q amplification, and shortened telomeres. Sequencing identified more IM patients with active Helicobacter pylori infection compared with histopathology (11%-27%). Several IMs exhibited hypermethylation at DNA methylation valleys; however, IMs generally lack intragenic hypomethylation signatures of advanced malignancy. IM patients with shortened telomeres and chromosomal alterations were associated with subsequent dysplasia or GC; conversely patients exhibiting normal-like epigenomic patterns were associated with regression.


Subject(s)
Gastric Mucosa/pathology , Helicobacter Infections/genetics , Metaplasia/genetics , Precancerous Conditions/genetics , Stomach Neoplasms/etiology , Adult , Aged , DNA Methylation , Disease Progression , Epigenomics , Female , Gastric Mucosa/microbiology , Genomics , Helicobacter Infections/microbiology , Humans , Male , Metaplasia/microbiology , Middle Aged , Precancerous Conditions/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology
10.
Cancer Lett ; 412: 208-215, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29061504

ABSTRACT

Recent gastric cancer clinical trials have aimed to establish the efficacy of combination therapy over monotherapy, however, the role for genomic biomarkers in these trials has remained largely unexplored. Here, using the NanoString expression platform, we analyzed 105 gastric tumors from a randomized phase III Japanese clinical trial (GC0301/TOP002) testing the efficacy of irinotecan plus S-1(IRI-S) versus S-1 therapy. We found that previously established proliferative subtype signatures, were associated with older patients (>65 years) and liver metastasis while mesenchymal subtype signatures were associated with younger patients (≤65 years) and peritoneal metastasis. Genes associated with tumor microenvironment (CD4, CD14, ADAMTS1, CCL5, CXCL12, CCL19), therapeutic implications (DPYD) and oncogenic signaling (Wnt5A, PTRF) were significantly associated with patient age, histology, tumor status, measurable lesions and metastasis. We identified Wnt5A downregulation as a candidate predictor of improved progression free survival (>8 weeks) in S-1 but not in IRI-S treatment. Although statistical significance was not achieved, mesenchymal subtype showed a trend for treatment interaction with IRI-S for efficacy. These findings highlight promising genomic markers that could be useful predictors of chemotherapy efficacy for better prognosis and survival outcome in gastric cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Stomach Neoplasms/drug therapy , Adult , Aged , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Drug Combinations , Female , Humans , Irinotecan , Liver Neoplasms/secondary , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Oxonic Acid/administration & dosage , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Tegafur/administration & dosage , Wnt-5a Protein/genetics
11.
Mol Cancer Ther ; 17(1): 232-242, 2018 01.
Article in English | MEDLINE | ID: mdl-28978722

ABSTRACT

Preclinical models of diffuse-type gastric cancer (DGC) that reliably predict clinical activity of novel compounds are lacking. To overcome the problem of poor tumor cellularity in DGC, we used cells from malignant ascites to establish DGC patient-derived xenograft (PDX) models that recapitulate the primary cancer. Cells in PDX model GAGA6 with FGFR2 amplification were sensitive to AZD4547, a potent FGFR inhibitor that is being clinically evaluated for FGFR-aberrant cancer types. Intermittent in vivo treatment of GAGA6 tumors with AZD4547 gave rise to PDX tumors with acquired resistance to AZD4547, GAGA6-R. Surprisingly, there were no mutations in the FGFR2 gene in GAGA6-R, negating gatekeeper mutations as a mechanism of drug resistance. Phosphorylation of FGFR2 and downstream signaling molecules AKT/PKB and MAPK/ERK remained inhibited by AZD4547. Further analysis of signaling pathways identified AKT-independent phosphorylation and inhibition of GSK3ß as a mechanism of drug resistance in GAGA6-R cells. Treatment of GAGA6-R cells with protein kinase C (PKC) inhibitor H7 in combination with AZD4547 led to dephosphorylation and activation of GSK3ß with concomitant downregulation of MCL-1 and BCL-XL. Combined treatment with AZD4547 and H7 in vitro synergistically enhanced cell death in GAGA6-R but not GAGA6 cells. Furthermore, midostaurin, a multikinase inhibitor with PKC-inhibiting activity, in part reversed resistance of GAGA6-R tumor to AZD4547 in vivo Our results suggest that upon challenge with FGFR inhibitors, FGFR2-amplified tumors that are highly dependent on FGFR2 signaling for survival rapidly develop resistance by switching to a PKC-mediated inhibition of GSK3ß to gain a survival advantage. Mol Cancer Ther; 17(1); 232-42. ©2017 AACR.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Stomach Neoplasms/genetics , Animals , Apoptosis , Cell Line, Tumor , Humans , Mice , Mice, Inbred NOD , Phosphorylation , Signal Transduction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Transfection
12.
Ann Surg ; 267(1): 114-121, 2018 Jan.
Article in English | MEDLINE | ID: mdl-27779515

ABSTRACT

OBJECTIVE: To establish the gene copy number status of receptor tyrosine kinase (RTK) and downstream signaling (DSS) genes genes in primary gastric cancer (primGC) and matched lymph node metastases (LNmet). BACKGROUND: Evidence suggests that coamplification between RTKs and DSSs and conversion between primGC and LNmet are associated with resistance to targeted therapy. METHODS: DNA from 237 Japanese primGC and 103 matched LNmet was analyzed using a newly developed multiplex ligation-dependent probe amplification (MLPA) probemix to investigate RTK (EGFR, HER2, FGFR2, and MET) and DSS (PIK3CA, KRAS, MYC, and CCNE1) gene copy number status. Results were compared between primGC and LNmet and related to clinicopathological data including survival. RESULTS: A total of 150 (63%) primGC had either RTK or DSS amplification. DSS coamplification was more frequent than RTK coamplification in primGC and LNmets. Moreover, 70 (30%) GC showed a disconcordant RTK and/or DSS gene copy number status between primGC and LNmet, most common was negative conversion for DSS genes (n=40 GC). The presence of RTK amplification in primGC was related to poorer survival in univariate analysis (P=0.04). CONCLUSIONS: This is the first and most comprehensive study in gastric cancer investigating the concordance between gene copy number status of targetable RTKs and downstream signaling oncogenes in primGC and LNmets. Future studies need to establish whether the relative high frequency of RTK and DSS coamplification and/or the relative high rate of negative conversion in LNmet can potentially explain recent failures of RTK targeted therapy in gastric cancer patients.


Subject(s)
Lymph Nodes/pathology , Receptor Protein-Tyrosine Kinases/genetics , Stomach Neoplasms/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Female , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Incidence , Japan/epidemiology , Lymphatic Metastasis/genetics , Male , Neoplasm Staging , Nucleic Acid Amplification Techniques , Receptor Protein-Tyrosine Kinases/metabolism , Retrospective Studies , Stomach Neoplasms/epidemiology , Stomach Neoplasms/secondary , Survival Rate/trends
13.
ESMO Open ; 1(1): e000009, 2016.
Article in English | MEDLINE | ID: mdl-27843583

ABSTRACT

BACKGROUND: Gene expression profiling has contributed greatly to cancer research. However, expression-driven biomarker discovery in metastatic gastric cancer (mGC) remains unclear. A gene expression profile predicting RAD001 response in refractory GC was explored in this study. METHODS: Total RNA isolated from 54 tumour specimens from patients with mGC, prior to RAD001 treatment, was analysed via the NanoString nCounter gene expression assay. This assay targeted 477 genes representing 10 different GC-related oncogenic signalling and molecular subtype-specific expression signatures. Gene expression profiles were correlated with patient clinicopathological variables. RESULTS: NanoString data confirmed similar gene expression profiles previously identified by microarray analysis. Signature I with 3 GC subtypes (mesenchymal, metabolic and proliferative) showed approximately 90% concordance where the mesenchymal and proliferative subtypes were significantly associated with signet ring cell carcinoma and the WHO classified tubular adenocarcinoma GC, respectively (p=0.042). Single-gene-level correlations with patient clinicopathological variables showed strong associations between FHL1 expression (mesenchymal subtype) and signet ring cell carcinoma, and NEK2, OIP5, PRC1, TPX2 expression (proliferative subtype) with tubular adenocarcinoma (adjusted p<0.05). Increased BRCA2 (p=0.040) and MMP9 (p=0.045) expression was significantly associated with RAD001 good response and longer progression-free survival outcome (BRCA2, p=0.012, HR 0.370 95% CI (0.171 to 0.800); MMP9, p=0.010, HR 0.359 95% CI (0.166 to 0.779)). In contrast, increased BTC (p=0.035) expression was significantly associated with RAD001 poor response and poor progression-free survival (p=0.031, HR 2.336 95% CI (1.079 to 5.059) by univariate Cox regression analysis. CONCLUSIONS: Microarray results are highly reproducible with NanoString nCounter gene expression profiling. Additionally, BRCA2 and MMP9 expression are potential predictive biomarkers for good response in RAD001-treated mGC.

14.
Sci Rep ; 6: 19552, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26790612

ABSTRACT

Dacomitinib, an irreversible pan-HER inhibitor, had shown modest clinical activity in squamous cell carcinoma of head and neck (SCCHN) patients. Therefore, validated predictive biomarkers are required to identify patients most likely to benefit from this therapeutic option. To characterize the genetic landscape of cisplatin-treated SCCHN genomes and identify potential predictive biomarkers for dacomitinib sensitivity, we performed whole exome sequencing on 18 cisplatin-resistant metastatic SCCHN tumors and their matched germline DNA. Platinum-based chemotherapy elevated the mutation rates of SCCHN compared to chemotherapy-naïve SCCHNs. Cisplatin-treated SCCHN genomes uniquely exhibited a novel mutational signature characterized by C:G to A:T transversions at CCR sequence contexts that may have arisen due to error-prone translesional synthesis. Somatic mutations in REV3L, the gene encoding the catalytic subunit of DNA polymerase ζ involved in translesional synthesis, are significantly enriched in a subset of patients who derived extended clinical benefit to dacomitinib (P = 0.04). Functional assays showed that loss-of-function of REV3L dramatically enhanced the sensitivity of SCCHN cells to dacomitinib by the loss of both translesion synthesis and homologous recombination pathways. Our data suggest that the 'platinum' mutational signature and inactivation of REV3L may inform treatment options in patients of recurrent SCCHN.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cisplatin/pharmacology , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Drug Resistance, Neoplasm/genetics , Exome , Head and Neck Neoplasms/genetics , High-Throughput Nucleotide Sequencing , Mutation , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Cisplatin/therapeutic use , DNA Mutational Analysis , Gene Silencing , Head and Neck Neoplasms/drug therapy , Humans , Quinazolinones/pharmacology , Quinazolinones/therapeutic use , RNA Interference , RNA, Small Interfering/genetics , Recombinational DNA Repair , Squamous Cell Carcinoma of Head and Neck
15.
Gut ; 65(12): 1960-1972, 2016 12.
Article in English | MEDLINE | ID: mdl-26338826

ABSTRACT

BACKGROUND: GI stromal tumours (GISTs) are clinically heterogenous exhibiting varying degrees of disease aggressiveness in individual patients. OBJECTIVES: We sought to identify genetic alterations associated with high-risk GIST, explore their molecular consequences, and test their utility as prognostic markers. DESIGNS: Exome sequencing of 18 GISTs was performed (9 patients with high-risk/metastatic and 5 patients with low/intermediate-risk), corresponding to 11 primary and 7 metastatic tumours. Candidate alterations were validated by prevalence screening in an independent patient cohort (n=120). Functional consequences of SETD2 mutations were investigated in primary tissues and cell lines. Transcriptomic profiles for 8 GISTs (4 SETD2 mutated, 4 SETD2 wild type) and DNA methylation profiles for 22 GISTs (10 SETD2 mutated, 12 SETD2 wild type) were analysed. Statistical associations between molecular, clinicopathological factors, and relapse-free survival were determined. RESULTS: High-risk GISTs harboured increased numbers of somatic mutations compared with low-risk GISTs (25.2 mutations/high-risk cases vs 6.8 mutations/low-risk cases; two sample t test p=3.1×10-5). Somatic alterations in the SETD2 histone modifier gene occurred in 3 out of 9 high-risk/metastatic cases but no low/intermediate-risk cases. Prevalence screening identified additional SETD2 mutations in 7 out of 80 high-risk/metastatic cases but no low/intermediate-risk cases (n=29). Combined, the frequency of SETD2 mutations was 11.2% (10/89) and 0% (0/34) in high-risk and low-risk GISTs respectively. SETD2 mutant GISTs exhibited decreased H3K36me3 expression while SETD2 silencing promoted DNA damage in GIST-T1 cells. In gastric GISTs, SETD2 mutations were associated with overexpression of HOXC cluster genes and a DNA methylation signature of hypomethylated heterochromatin. Gastric GISTs with SETD2 mutations, or GISTs with hypomethylated heterochromatin, showed significantly shorter relapse-free survival on univariate analysis (log rank p=4.1×10-5). CONCLUSIONS: Our data suggest that SETD2 is a novel GIST tumour suppressor gene associated with disease progression. Assessing SETD2 genetic status and SETD2-associated epigenomic phenotypes may guide risk stratification and provide insights into mechanisms of GIST clinical aggressiveness.


Subject(s)
Biomarkers, Tumor/genetics , Gastrointestinal Stromal Tumors/genetics , Histone-Lysine N-Methyltransferase/genetics , Mutation, Missense , Case-Control Studies , Codon, Nonsense/genetics , DNA Methylation/genetics , Exome/genetics , Gastrointestinal Stromal Tumors/epidemiology , Gastrointestinal Stromal Tumors/pathology , Histones/genetics , Humans , Mutation, Missense/genetics , Neoplasm Invasiveness , Phenotype , Prevalence , Prognosis , Severity of Illness Index , Singapore/epidemiology
16.
Gut ; 64(5): 707-19, 2015 May.
Article in English | MEDLINE | ID: mdl-25053715

ABSTRACT

OBJECTIVE: Gastric cancer (GC) is a deadly malignancy for which new therapeutic strategies are needed. Three transcription factors, KLF5, GATA4 and GATA6, have been previously reported to exhibit genomic amplification in GC. We sought to validate these findings, investigate how these factors function to promote GC, and identify potential treatment strategies for GCs harbouring these amplifications. DESIGN: KLF5, GATA4 and GATA6 copy number and gene expression was examined in multiple GC cohorts. Chromatin immunoprecipitation with DNA sequencing was used to identify KLF5/GATA4/GATA6 genomic binding sites in GC cell lines, and integrated with transcriptomics to highlight direct target genes. Phenotypical assays were conducted to assess the function of these factors in GC cell lines and xenografts in nude mice. RESULTS: KLF5, GATA4 and GATA6 amplifications were confirmed in independent GC cohorts. Although factor amplifications occurred in distinct sets of GCs, they exhibited significant mRNA coexpression in primary GCs, consistent with KLF5/GATA4/GATA6 cross-regulation. Chromatin immunoprecipitation with DNA sequencing revealed a large number of genomic sites co-occupied by KLF5 and GATA4/GATA6, primarily located at gene promoters and exhibiting higher binding strengths. KLF5 physically interacted with GATA factors, supporting KLF5/GATA4/GATA6 cooperative regulation on co-occupied genes. Depletion and overexpression of these factors, singly or in combination, reduced and promoted cancer proliferation, respectively, in vitro and in vivo. Among the KLF5/GATA4/GATA6 direct target genes relevant for cancer development, one target gene, HNF4α, was also required for GC proliferation and could be targeted by the antidiabetic drug metformin, revealing a therapeutic opportunity for KLF5/GATA4/GATA6 amplified GCs. CONCLUSIONS: KLF5/GATA4/GATA6 may promote GC development by engaging in mutual crosstalk, collaborating to maintain a pro-oncogenic transcriptional regulatory network in GC cells.


Subject(s)
GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic/genetics , Kruppel-Like Transcription Factors/genetics , Stomach Neoplasms/genetics , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , GATA4 Transcription Factor/biosynthesis , GATA6 Transcription Factor/biosynthesis , Gene Expression Profiling/methods , Gene Silencing , Genetic Predisposition to Disease , Heterografts , Humans , Kruppel-Like Transcription Factors/biosynthesis , Mice, Nude , Neoplasm Transplantation , Oncogenes/genetics , Promoter Regions, Genetic , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Cells, Cultured
17.
Cancer Lett ; 353(2): 167-75, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25086186

ABSTRACT

Gastric cancer (GC) is a major cause of global cancer mortality. Previous genomic studies have reported that several RTK-RAS pathway components are amplified in GC, with individual tumours often amplifying one component and not others ("mutual exclusivity"). Here, we sought to validate these findings for three RTK/RAS components (FGFR2, HER2, KRAS) using fluorescence in situ hybridisation (FISH) on a series of gastric tumours, cell lines and patient-derived xenografts. Applying dual-colour FISH on 137 gastric tumours (89 FFPE surgical resections and 48 diagnostic biopsies), we observed FGFR2 amplification in 7.3% and HER2 amplification in 2.2% of GCs. GCs exhibiting FGFR2 amplification were associated with high tumour grade (p = 0.034). In FISH positive tumours, striking differences in copy number levels between cancer cells in the same tumour were observed, suggesting intra-tumour heterogeneity. Using a multicolour FISH assay allowing simultaneous detection of FGFR2, HER2, and KRAS amplifications, we confirmed that these components exhibited a mutually exclusive pattern of gene amplification across patients. The FISH data were also strongly correlated with Q-PCR levels and at the protein level by immunohistochemistry. Our data confirm that RTK/RAS components are mutually exclusively amplified in GC, and demonstrate the feasibility of identifying multiple aneuploidies using a single FISH assay. Application of this assay to GC samples, particularly diagnostic biopsies, may facilitate enrollment of GC patients into clinical trials evaluating RTK/RAS directed therapies. However, the presence of intra-tumour heterogeneity may require multiple biopsy samples to be obtained per patient before a definitive diagnosis can be attained.


Subject(s)
Gene Amplification , Proto-Oncogene Proteins/genetics , Receptor, ErbB-2/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Stomach Neoplasms/genetics , ras Proteins/genetics , Aged , Biomarkers, Tumor/genetics , Female , Gene Dosage , Genetic Association Studies , Humans , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Male , Middle Aged , Proto-Oncogene Proteins p21(ras) , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology
18.
PLoS One ; 9(3): e90133, 2014.
Article in English | MEDLINE | ID: mdl-24598828

ABSTRACT

Despite the benefits from adjuvant chemotherapy or chemoradiotherapy, approximately one-third of stage II gastric cancer (GC) patients developed recurrences. The aim of this study was to develop and validate a prognostic algorithm for gastric cancer (GCPS) that can robustly identify high-risk group for recurrence among stage II patients. A multi-step gene expression profiling study was conducted. First, a microarray gene expression profiling of archived paraffin-embedded tumor blocks was used to identify candidate prognostic genes (N=432). Second, a focused gene expression assay including prognostic genes was used to develop a robust clinical assay (GCPS) in stage II patients from the same cohort (N=186). Third, a predefined cut off for the GCPS was validated using an independent stage II cohort (N=216). The GCPS was validated in another set with stage II GC who underwent surgery without adjuvant treatment (N=300). GCPS was developed by summing the product of Cox regression coefficients and normalized expression levels of 8 genes (LAMP5, CDC25B, CDK1, CLIP4, LTB4R2, MATN3, NOX4, TFDP1). A prospectively defined cut-point for GCPS classified 22.7% of validation cohort treated with chemoradiotherapy (N=216) as high-risk group with 5-year recurrence rate of 58.6% compared to 85.4% in the low risk group (hazard ratio for recurrence=3.16, p=0.00004). GCPS also identified high-risk group among stage II patients treated with surgery only (hazard ratio=1.77, p=0.0053).


Subject(s)
Adenocarcinoma/metabolism , Gene Expression Profiling , Neoplasm Recurrence, Local/metabolism , Stomach Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/surgery , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chemoradiotherapy, Adjuvant , Disease-Free Survival , Humans , Kaplan-Meier Estimate , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Matrilin Proteins/genetics , Matrilin Proteins/metabolism , Membrane Proteins , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/prevention & control , Oligonucleotide Array Sequence Analysis , Prognosis , Proportional Hazards Models , Risk , Stomach Neoplasms/genetics , Stomach Neoplasms/surgery , Transcriptome
19.
Gastroenterology ; 145(3): 554-65, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23684942

ABSTRACT

BACKGROUND & AIMS: Almost all gastric cancers are adenocarcinomas, which have considerable heterogeneity among patients. We sought to identify subtypes of gastric adenocarcinomas with particular biological properties and responses to chemotherapy and targeted agents. METHODS: We compared gene expression patterns among 248 gastric tumors; using a robust method of unsupervised clustering, consensus hierarchical clustering with iterative feature selection, we identified 3 major subtypes. We developed a classifier for these subtypes and validated it in 70 tumors from a different population. We identified distinct genomic and epigenomic properties of the subtypes. We determined drug sensitivities of the subtypes in primary tumors using clinical survival data, and in cell lines through high-throughput drug screening. RESULTS: We identified 3 subtypes of gastric adenocarcinoma: proliferative, metabolic, and mesenchymal. Tumors of the proliferative subtype had high levels of genomic instability, TP53 mutations, and DNA hypomethylation. Cancer cells of the metabolic subtype were more sensitive to 5-fluorouracil than the other subtypes. Furthermore, in 2 independent groups of patients, those with tumors of the metabolic subtype appeared to have greater benefits with 5-fluorouracil treatment. Tumors of the mesenchymal subtype contain cells with features of cancer stem cells, and cell lines of this subtype are particularly sensitive to phosphatidylinositol 3-kinase-AKT-mTOR inhibitors in vitro. CONCLUSIONS: Based on gene expression patterns, we classified gastric cancers into 3 subtypes, and validated these in an independent set of tumors. The subgroups have differences in molecular and genetic features and response to therapy; this information might be used to select specific treatment approaches for patients with gastric cancer.


Subject(s)
Adenocarcinoma/classification , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Fluorouracil/therapeutic use , Gene Expression Regulation, Neoplastic , Phosphoinositide-3 Kinase Inhibitors , Stomach Neoplasms/classification , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Aged , Bayes Theorem , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cluster Analysis , Genetic Association Studies , Humans , Middle Aged , Models, Statistical , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Regression Analysis , Retrospective Studies , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Analysis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Treatment Outcome
20.
Gut ; 61(5): 673-84, 2012 May.
Article in English | MEDLINE | ID: mdl-22315472

ABSTRACT

OBJECTIVE: Gastric cancer is a major gastrointestinal malignancy for which targeted therapies are emerging as treatment options. This study sought to identify the most prevalent molecular targets in gastric cancer and to elucidate systematic patterns of exclusivity and co-occurrence among these targets, through comprehensive genomic analysis of a large panel of gastric cancers. DESIGN: Using high-resolution single nucleotide polymorphism arrays, copy number alterations were profiled in a panel of 233 gastric cancers (193 primary tumours, 40 cell lines) and 98 primary matched gastric non-malignant samples. For selected alterations, their impact on gene expression and clinical outcome were evaluated. RESULTS: 22 recurrent focal alterations (13 amplifications and nine deletions) were identified. These included both known targets (FGFR2, ERBB2) and also novel genes in gastric cancer (KLF5, GATA6). Receptor tyrosine kinase (RTK)/RAS alterations were found to be frequent in gastric cancer. This study also demonstrates, for the first time, that these alterations occur in a mutually exclusive fashion, with KRAS gene amplifications highlighting a clinically relevant but previously underappreciated gastric cancer subgroup. FGFR2-amplified gastric cancers were also shown to be sensitive to dovitinib, an orally bioavailable FGFR/VEGFR targeting agent, potentially representing a subtype-specific therapy for FGFR2-amplified gastric cancers. CONCLUSION: The study demonstrates the existence of five distinct gastric cancer patient subgroups, defined by the signature genomic alterations FGFR2 (9% of tumours), KRAS (9%), EGFR (8%), ERBB2 (7%) and MET (4%). Collectively, these subgroups suggest that at least 37% of gastric cancer patients may be potentially treatable by RTK/RAS directed therapies.


Subject(s)
Gene Amplification , Gene Deletion , Polymorphism, Single Nucleotide , Stomach Neoplasms/genetics , Antineoplastic Agents/therapeutic use , ErbB Receptors/genetics , Genetic Markers , Molecular Targeted Therapy , Oligonucleotide Array Sequence Analysis , Proportional Hazards Models , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins p21(ras) , Receptor, ErbB-2/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/mortality , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...