Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Front Oncol ; 14: 1272432, 2024.
Article in English | MEDLINE | ID: mdl-38939336

ABSTRACT

Introduction: Field cancerization is suggested to arise from imbalanced differentiation in individual basal progenitor cells leading to clonal expansion of mutant cells that eventually replace the epithelium, although without evidence. Methods: We performed deep sequencing analyses to characterize the genomic and transcriptomic landscapes of field change in two patients with synchronous aerodigestive tract tumors. Results: Our data support the emergence of numerous genetic alterations in cancer-associated genes but refutes the hypothesis that founder mutation(s) underpin this phenomenon. Mutational signature analysis identified defective homologous recombination as a common underlying mutational process unique to synchronous tumors. Discussion: Our analyses suggest a common etiologic factor defined by mutational signatures and/or transcriptomic convergence, which could provide a therapeutic opportunity.

2.
Clin Cancer Res ; 30(7): 1223-1225, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38252056

ABSTRACT

Distinguishing low- versus high-risk HPV-associated oropharyngeal squamous cell carcinoma (OPSCC) is pivotal for tailoring treatment. Liquid biopsy, measuring cell-free HPV-DNA in serum and saliva, assesses treatment response and early-recurrence risk. Postoperative lymphatic fluid may better guide future adjuvant therapy decisions due to its proximity to primary lesions and lymph nodes. See related article by Earland et al., p. 1409.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Oropharyngeal Neoplasms , Papillomavirus Infections , Humans , Squamous Cell Carcinoma of Head and Neck/therapy , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/therapy , Carcinoma, Squamous Cell/pathology , Oropharyngeal Neoplasms/diagnosis , Oropharyngeal Neoplasms/therapy , Oropharyngeal Neoplasms/pathology , Papillomavirus Infections/complications , Papillomavirus Infections/diagnosis , Papillomavirus Infections/pathology , Papillomaviridae/genetics
3.
Ann Surg Open ; 4(4): e339, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38144489

ABSTRACT

Objective: To examine the association between the performance of mapping biopsies and surgical outcomes postexcision of extramammary Paget's disease (EMPD). Background: Primary EMPD is a rare entity associated with poorly defined surgical margins and difficult-to-access sites of lesions. Surgical resection with clear margins remains the preferred management method. The use of mapping biopsies might be beneficial, particularly in lowering disease recurrence. Methods: Available literature was reviewed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses methodology before a fixed-effect meta-analysis was performed to identify the presence of a correlation between performing mapping biopsies and positive margins on permanent sections as well as disease-free survival. Additional study results not included in the quantitative assessment were qualitatively assessed and reported. Results: A total of 12 studies were shortlisted for final analysis. 294 patients who underwent mapping biopsies and 48 patients who did not undergo mapping biopsies were included in the assessment. Forest plot analysis revealed a pooled rate ratio of 0.50 (95% CI, 0.32-0.77) in the prevalence of positive margins in patients with mapping biopsies performed as compared to patients without. The pooled rate ratio of the prevalence of disease-free survival in patients with mapping biopsies performed as compared to patients without was 1.38 (95% CI, 1.03-1.84). Qualitative assessment of the remaining selected studies revealed equivocal results. Conclusions: Mapping biopsies are able to improve EMPD surgical excision outcomes but given the rarity of the disease and heterogeneity of mapping biopsy procedures, further confirmation with randomized controlled trials or a larger patient pool is necessary.

4.
Front Med (Lausanne) ; 10: 1281843, 2023.
Article in English | MEDLINE | ID: mdl-38105890

ABSTRACT

Introduction: Prehabilitation, which involves improving a patient's physical and psychological condition before surgery, has shown potential benefits but has yet to be extensively studied from an economic perspective. To address this gap, a systematic review was conducted to summarize existing economic evaluations of prehabilitation interventions. Methods: The PRISMA Protocols 2015 checklist was followed. Over 16,000 manuscripts were reviewed, and 99 reports on preoperative interventions and screening tests were identified, of which 12 studies were included in this analysis. The costs are expressed in Pounds (GBP, £) and adjusted for inflation to December 2022. Results: The studies were conducted in Western countries, focusing on specific surgical subspecialties. While the interventions and study designs varied, most studies demonstrated cost savings in the intervention group compared to the control group. Additionally, all cost-effectiveness analysis studies favored the intervention group. However, the review also identified several limitations. Many studies had a moderate or high risk of bias, and critical information such as time horizons and discount rates were often missing. Important components like heterogeneity, distributional effects, and uncertainty were frequently lacking as well. The misclassification of economic evaluation types highlighted a lack of knowledge among physicians in prehabilitation research. Conclusion: This review reveals a lack of robust evidence regarding the economics of prehabilitation programs for surgical patients. This suggests a need for further research with rigorous methods and accurate definitions.

5.
Cell Rep Med ; 3(2): 100526, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35243423

ABSTRACT

Peritoneal carcinomatosis (PC) present a ubiquitous clinical conundrum in all intra-abdominal malignancies. Via functional and transcriptomic experiments of ascites-treated PC cells, we identify STAT3 as a key signaling pathway. Integrative analysis of publicly available databases and correlation with clinical cohorts (n = 7,359) reveal putative clinically significant activating ligands of STAT3 signaling. We further validate a 3-biomarker prognostic panel in ascites independent of clinical covariates in a prospective study (n = 149). Via single-cell sequencing experiments, we uncover that PAI-1, a key component of the prognostic biomarker panel, is largely secreted by fibroblasts and mesothelial cells. Molecular stratification of ascites using PAI-1 levels and STAT3 activation in ascites-treated cells highlight a therapeutic opportunity based on a phenomenon of paracrine addiction. These results are recapitulated in patient-derived ascites-dependent xenografts. Here, we demonstrate therapeutic proof of concept of direct ligand inhibition of a prognostic target within an enclosed biological space.


Subject(s)
Peritoneal Neoplasms , Animals , Ascites , Disease Models, Animal , Humans , Ligands , Mice , Plasminogen Activator Inhibitor 1/genetics , Prospective Studies
6.
Semin Thorac Cardiovasc Surg ; 33(3): 750-759, 2021.
Article in English | MEDLINE | ID: mdl-33181310

ABSTRACT

HbA1C's predictive value for postoperative complications in cardiac surgery has been mixed. Studies did not account for HbA1C being over-read in anemic patients. This study proposes a novel way of using a ratio of HbA1C over hemoglobin (HH ratio). Retrospective recruitment of patients undergoing cardiac surgery was done with ethics approval. The primary objective of our study is to look for the correlation of HH ratio with 90-day (short-term) and 1-year (long-term) mortality. The secondary objective is to investigate its association with other adverse events. Statistical analysis was done using multivariable regressions and Cox proportional hazard models. Of the 974 patients recruited, 618 had a HH Ratio<0.5, 284 between 0.5-0.7 and 72 had the ratio >0.7. HH ratio of 0.5-0.7 and >0.7 was associated with 90-day mortality (HR 5.12, P = 0.033 and HR 7.25, P= 0.048 respectively) and 1-year mortality (HR 4.53, P = 0.028 and HR 9.20, P = 0.022 respectively). The higher HH ratio groups were also associated with increased length of stay (hours) in the intensive care unit (P < 0.001) and renal complications (P < 0.001). Our study showed a positive association of HH ratio with 90-day and 1-year mortality and postoperative adverse outcomes in patients undergoing cardiac surgery. The HH ratio has the potential to be a new perioperative target.


Subject(s)
Anemia , Cardiac Surgical Procedures , Anemia/diagnosis , Anemia/etiology , Cardiac Surgical Procedures/adverse effects , Glycated Hemoglobin , Hemoglobins , Humans , Postoperative Complications/etiology , Retrospective Studies , Risk Factors
7.
Oral Oncol ; 111: 105035, 2020 12.
Article in English | MEDLINE | ID: mdl-33091845

ABSTRACT

OBJECTIVES: We have previously identified and validated a panel of molecular prognostic markers (ATP13A3, SSR3, and ANO1) for Head and Neck Squamous Cell Carcinoma (HNSCC). The aim of this study was to investigate the consequence of ATP13A3 dysregulation on signaling pathways, to aid in formulating a therapeutic strategy targeting ATP13A3-overexpressing HNSCC. MATERIALS AND METHODS: Gene Set Enrichment Analysis (GSEA) was performed on HNSCC microarray expression data (Internal local dataset [n = 92], TCGA [n = 232], EMBL [n = 81]) to identify pathways associated with high expression of ATP13A3. Validation was performed using immunohistochemistry (IHC) on tissue microarrays (TMAs) of head and neck cancers (n = 333), staining for ATP13A3 and phosphorylated Aurora kinase A (phospho-T288). Short interfering RNA was used to knockdown ATP13A3 expression in patient derived HNSCC cell lines. Protein expression of ATP13A3 and Aurora kinase A was then assessed by immunoblotting. RESULTS: GSEA identified Aurora kinase pathway to be associated with high expression of ATP13A3 (p = 0.026). The Aurora kinase pathway was also associated with a trend towards poor prognosis and tumor aggressiveness (p = 0.086, 0.094, respectively). Furthermore, the immunohistochemical staining results revealed a significant association between Aurora kinase activity and high ATP13A3 expression (p < 0.001). Knockdown of ATP13A3 in human head and neck cell lines showed decrease in Aurora kinase A levels. CONCLUSION: Tumors with high ATP13A3 are associated with high Aurora kinase activity. This suggests a potential therapeutic role of Aurora kinase inhibitors in a subset of poor prognosis HNSCC patients with overexpression of ATP13A3.


Subject(s)
Adenosine Triphosphatases/metabolism , Aurora Kinase A/metabolism , Head and Neck Neoplasms/metabolism , Membrane Transport Proteins/metabolism , Protein Kinase Inhibitors/therapeutic use , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/metabolism , Adenosine Triphosphatases/genetics , Aurora Kinase A/antagonists & inhibitors , Cell Line, Tumor , Female , Gene Silencing , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Male , Membrane Transport Proteins/genetics , Molecular Targeted Therapy/methods , Prognosis , RNA, Small Interfering , Signal Transduction/genetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Tissue Array Analysis
8.
Sci Rep ; 10(1): 682, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959771

ABSTRACT

Generation of large amounts of genomic data is now feasible and cost-effective with improvements in next generation sequencing (NGS) technology. Ribonucleic acid sequencing (RNA-Seq) is becoming the preferred method for comprehensively characterising global transcriptome activity. Unique to cytoreductive surgery (CRS), multiple spatially discrete tumour specimens could be systematically harvested for genomic analysis. To facilitate such downstream analyses, laser capture microdissection (LCM) could be utilized to obtain pure cell populations. The aim of this protocol study was to develop a methodology to obtain high-quality expression data from matched primary tumours and metastases by utilizing LCM to isolate pure cellular populations. We demonstrate an optimized LCM protocol which reproducibly delivered intact RNA used for RNA sequencing and quantitative polymerase chain reaction (qPCR). After pathologic annotation of normal epithelial, tumour and stromal components, LCM coupled with cDNA library generation provided for successful RNA sequencing. To illustrate our framework's potential to identify targets that would otherwise be missed with conventional bulk tumour sequencing, we performed qPCR and immunohistochemical technical validation to show that the genes identified were truly expressed only in certain sub-components. This study suggests that the combination of matched tissue specimens with tissue microdissection and NGS provides a viable platform to unmask hidden biomarkers and provides insight into tumour biology at a higher resolution.


Subject(s)
Colorectal Neoplasms/surgery , Gene Expression Profiling/methods , Krukenberg Tumor/surgery , Laser Capture Microdissection/methods , Ovarian Neoplasms/surgery , Colorectal Neoplasms/genetics , Colorectal Neoplasms/secondary , Female , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Krukenberg Tumor/genetics , Ovarian Neoplasms/genetics , Sequence Analysis, RNA , Specimen Handling , Workflow
9.
Asian J Urol ; 5(3): 194-198, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29988907

ABSTRACT

OBJECTIVE: Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) are increasingly being used to treat peritoneal malignancies. Urological resections and reconstruction (URR) are occasionally performed during the surgery. We aim to evaluate the impact of these procedures on peri-operative outcomes of CRS and HIPEC patients. METHODS: A retrospective review of a prospectively maintained database of all patients who underwent CRS-HIPEC from April 2001 to February 2016 was performed. Outcomes between patients who had surgery involving, and not involving URR were compared. Primary outcomes were the rate of major complications and the duration of stay in the intensive care unit (ICU) and hospital. Secondary outcomes were that of overall survival (OS) and prognostic factors that would indicate a need for URR. RESULTS: A total of 214 CRS-HIPEC were performed, 21 of which involved a URR. Baseline clinical characteristics did not vary between the groups (URR vs. No URR). Urological resections comprised of 52% bladder resections, 24% ureteric resections, and 24% involving both bladder and ureteric resections. All bladder defects were closed primarily while ureteric reconstructions consisted of two end-to-end anastomoses, one ureto-uretostomy, five direct implantations into the bladder and three boari flaps. URR were more frequently required in patients with colorectal peritoneal disease (p = 0.029), but was not associated with previous pelvic surgery (76% vs. 54%, p = 0.065). Patients with URR did not suffer more serious complications (14% vs. 24%, p = 0.42). ICU (2.2 days vs. 1.4 days, p = 0.51) and hospital stays (18 days vs. 25 days, p = 0.094) were not significantly affected. Undergoing a URR did not affect OS (p = 0.99), but was associated with increased operation time (570 min vs. 490 min, p = 0.046). CONCLUSION: While concomitant URR were associated with an increase in operation time, there were no significant differences in postoperative complications or OS. Patients with colorectal peritoneal metastases are more likely to require a URR compared to other primary tumours, and needs to be considered during pre-operative planning.

10.
Pleura Peritoneum ; 3(4): 20180122, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30911666

ABSTRACT

BACKGROUND: Cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) is routinely used for selected patients with peritoneal metastasis, but can be associated with high complication rates, prolonged hospital stay, and mortality. Our objective was to determine the learning curve of CRS/HIPEC in our institution, representing the largest Asian cohort to date. METHODS: A total of 200 consecutive patients with peritoneal metastasis treated with CRS/HIPEC between 2001 and 2016 were grouped into four cohorts of 50 patients and studied. Primary outcomes were severe morbidity (Clavien-Dindo III-V), procedure-related mortality, and duration of ICU and hospital stays. Secondary outcome was duration of surgery. RESULTS: Median age was 53 years (10-75). There was no significant age, sex, or histology difference across cohorts. Rates of severe morbidity (23 %), and 60 day inpatient mortality (0.5 %) were comparable to previously reported data. Decreases in rates of serious morbidity, (34 %, 30 %, 12 %, 14 %, p<0.01) and duration of total hospital stay (14, 16, 13, 12 days, p=0.041) were seen across consecutive cohorts. Operation time decreased significantly after the first cohort (10, 7.8, 7.8, 7.2 h, p<0.01), despite increase in average PCI score after the first cohort (8, 14, 12, 13, p=0.063). CONCLUSIONS: Whilst 50 cases were adequate for procedural familiarity and decreased average operation time, significant improvement in rate of serious morbidity was observed after 100 operations. We demonstrate a novel biphasic nature to the learning curve, reflecting initial training in which technical competence is achieved, followed by a subsequent period characterized by increasingly complex cases (higher PCI score) and finally refinement of patient selection.

11.
Oncogene ; 37(10): 1340-1353, 2018 03.
Article in English | MEDLINE | ID: mdl-29255247

ABSTRACT

Treatment failure in solid tumors occurs due to the survival of specific subpopulations of cells that possess tumor-initiating (TIC) phenotypes. Studies have implicated G protein-coupled-receptors (GPCRs) in cancer progression and the acquisition of TIC phenotypes. Many of the implicated GPCRs signal through the G protein GNA13. In this study, we demonstrate that GNA13 is upregulated in many solid tumors and impacts survival and metastases in patients. GNA13 levels modulate drug resistance and TIC-like phenotypes in patient-derived head and neck squamous cell carcinoma (HNSCC) cells in vitro and in vivo. Blockade of GNA13 expression, or of select downstream pathways, using small-molecule inhibitors abrogates GNA13-induced TIC phenotypes, rendering cells vulnerable to standard-of-care cytotoxic therapies. Taken together, these data indicate that GNA13 expression is a potential prognostic biomarker for tumor progression, and that interfering with GNA13-induced signaling provides a novel strategy to block TICs and drug resistance in HNSCCs.


Subject(s)
Cell Transformation, Neoplastic/genetics , Drug Resistance, Neoplasm/genetics , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Transformation, Neoplastic/drug effects , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Phenotype , Signal Transduction/drug effects , Signal Transduction/genetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Cells, Cultured
12.
Oncotarget ; 8(45): 79556-79566, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-29108335

ABSTRACT

BACKGROUND: Current management of head and neck squamous cell carcinoma (HNSCC) depends on tumor staging. Despite refinements in clinical staging algorithms, outcomes remain unchanged for the last two decades. In this study, we set out to identify a small, clinically applicable molecular panel to aid prognostication of patients with HNSCC. MATERIALS AND METHODS: Data from The Cancer Genome Atlas (TCGA) was used to derive copy number aberrations and expression changes to identify putative prognostic genes. To account for cross entity relevance of the biomarkers, HNSCC (n = 276), breast (n = 808) and lung cancer (n = 282) datasets were used to identify robust and reproducible markers with prognostic potential. Validation was performed using immunohistochemistry (IHC) on tissue microarrays of an independent cohort of HNSCC (n = 333). FINDINGS: Using GISTIC algorithm together with gene expression analysis, we identified six putative prognostic genes in at least two out of three cancers analyzed, of which four were successfully optimized for automated IHC. Of these, three were successfully validated; each molecular target being significantly prognostic on univariate analysis. Patients were differentially segregated into four prognostic groups based on the number of genes dysregulated (p < 0.001). The IHC panel remained an independent predictor of survival after adjusting for known survival covariates including clinical staging criteria in a multivariate Cox regression model (p < 0.001). . INTERPRETATION: We have identified and validated a clinically applicable IHC biomarker panel that is independently associated with overall survival. This panel is readily applicable, serving as a useful adjunct to current staging systems and provides novel targets for future therapeutic strategies.

13.
J Natl Cancer Inst ; 106(5)2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24777112

ABSTRACT

BACKGROUND: Many prognostic biomarkers have been proposed recently. However, there is a lack of therapeutic strategies exploiting novel prognostic biomarkers. We aimed to propose therapeutic options in patients with overexpression of TRIM44, a recently identified prognostic gene. METHODS: Genomic and transcriptomic data of epithelial cancers (n = 1932), breast cancers (BCs; n = 1980) and esophago-gastric cancers (EGCs; n = 163) were used to identify genomic aberrations driving TRIM44 overexpression. The driver gene status of TRIM44 was determined using a small interfering RNA (siRNA) screen of the 11p13 amplicon. Integrative analysis was applied across multiple datasets to identify pathway activation and potential therapeutic strategies. Validation of the in silico findings were performed using in vitro assays, xenografts, and patient samples (n = 160). RESULTS: TRIM44 overexpression results from genomic amplification in 16.1% of epithelial cancers, including 8.1% of EGCs and 6.1% of BCs. This was confirmed using fluorescent in situ hybridization. The siRNA screen confirmed TRIM44 to be a driver of the amplicon. In silico analysis revealed an association between TRIM44 and mTOR signalling, supported by a decrease in mTOR signalling after siRNA knockdown of TRIM44 in cell lines and colocalization of TRIM44 and p-mTOR in patient samples. In vitro inhibition studies using an mTOR inhibitor (everolimus) decreased cell viability in two TRIM44-amplified cells lines by 88% and 70% compared with 35% in the control cell line. These findings were recapitulated in xenograft models. CONCLUSIONS: Genomic amplification drives TRIM44 overexpression in EGCs and BCs. Targeting the mTOR pathway provides a potential therapeutic option for TRIM44-amplified tumors.


Subject(s)
Carrier Proteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Animals , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/biosynthesis , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Gene Amplification , Gene Knockdown Techniques , Heterografts , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred BALB C , Molecular Targeted Therapy , Neoplasm Staging , Neoplasms/metabolism , Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tripartite Motif Proteins
14.
Gut ; 62(10): 1415-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-22773546

ABSTRACT

OBJECTIVE: The success of personalised therapy depends on identification and inhibition of the oncogene(s) on which that tumour is dependent. We aimed to determine whether a receptor tyrosine kinase (RTK) array could be used to select the most effective therapeutic strategies in molecularly heterogeneous oesophago-gastric adenocarcinomas. DESIGN: Gene expression profiling from oesophago-gastric tumours (n=75) and preinvasive stages (n=57) identified the active signalling pathways, which was confirmed using immunohistochemistry (n=434). RTK arrays on a cell line panel (n=14) determined therapeutic targets for in vitro cytotoxic testing. Feasibility of this personalised approach was tested in tumour samples (n=46). RESULTS: MAPK was the most frequently activated pathway (32/75 samples (42.7%)) with progressive enrichment in preinvasive disease stages (p<0.05) and ERK phosphorylation in 148/434 (34.3%) independent samples. Cell lines displayed a range of RTK activation profiles. When no RTKs were activated, tyrosine kinase inhibitors (TKIs) and a Mek inhibitor were not useful (MKN1). In lines with a dominant phosphorylated RTK (OE19, MKN45 and KATOIII), selection of this TKI or Mek in nM concentrations induced cytotoxicity and inhibited Erk and Akt phosphorylation. In cells lines with complex activation profiles (HSC39 and OE33), a combination of TKIs or Mek inhibition (in nM concentrations) was necessary for cytotoxicity and inhibition of Erk and Akt phosphorylation. Human tumours demonstrated diverse activation profiles and 65% of cases had two or more active RTKs. CONCLUSIONS: The MAPK pathway is commonly activated in oesophago-gastric cancer following activation of a variety of RTKs. Molecular phenotyping can inform a rational choice of targeted therapy.


Subject(s)
Esophageal Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Barrett Esophagus/drug therapy , Barrett Esophagus/genetics , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Biomarkers, Tumor/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Discovery/methods , Enzyme Activation/drug effects , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Feasibility Studies , Female , Gene Expression Profiling/methods , Humans , MAP Kinase Signaling System/physiology , Male , Middle Aged , Neoplasm Staging , Phosphorylation/drug effects , Precancerous Conditions/drug therapy , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Precision Medicine/methods , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
15.
Clin Cancer Res ; 19(4): 878-88, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23243219

ABSTRACT

PURPOSE: Endoscopic surveillance of Barrett's esophagus is problematic because dysplasia/early-stage neoplasia is frequently invisible and likely to be missed because of sampling bias. Molecular abnormalities may be more diffuse than dysplasia. The aim was therefore to test whether DNA methylation, especially on imprinted and X-chromosome genes, is able to detect dysplasia/early-stage neoplasia. EXPERIMENTAL DESIGN: 27K methylation arrays were used to find genes best able to differentiate between 22 Barrett's esophagus and 24 esophageal adenocarcinoma (EAC) samples. These were validated using pyrosequencing on a retrospective cohort (60 Barrett's esophagus, 36 dysplastic, and 90 EAC) and then in a prospective multicenter study (98 Barrett's esophagus patients, including 28 dysplastic and 9 early EAC) designed to utilize biomarkers to stratify patients according to their prevalent dysplasia/EAC status. RESULTS: Genes (23%) on the array, including 7% of X-linked and 69% of imprinted genes, have shown statistically significant changes in methylation in EAC versus Barrett's esophagus (Wilcoxon P < 0.05). 6/7 selected candidate genes were successfully internally (Pearson's P < 0.01) and externally validated (ANOVA P < 0.001). Four genes (SLC22A18, PIGR, GJA12, and RIN2) showed the greatest area under curve (0.988) to distinguish between Barrett's esophagus and dysplasia/EAC in the retrospective cohort. This methylation panel was able to stratify patients from the prospective cohort into three risk groups based on the number of genes methylated (low risk: <2 genes, intermediate: 2, and high: >2). CONCLUSION: Widespread DNA methylation changes were observed in Barrett's carcinogenesis including ≈70% of known imprinted genes. A four-gene methylation panel stratified patients with Barrett's esophagus into three risk groups with potential clinical utility.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , DNA Methylation/genetics , Genomic Imprinting , Adenocarcinoma/pathology , Barrett Esophagus/pathology , Genes, X-Linked , Humans , Neoplasm Staging , Neoplasms/genetics , Neoplasms/pathology , Risk Factors
16.
Carcinogenesis ; 31(9): 1669-75, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20584750

ABSTRACT

Nitric oxide (NO) has been shown to induce double strand DNA breaks in Barrett's oesophagus (BO) and in other cancers has a role in invasion. The specific aims of this study were to investigate whether NO can induce invasion in cells representative of different stages of Barrett's progression and to determine possible underlying mechanisms. Physiological concentrations of NO that mimic luminal production of NO from dietary sources enhanced invasion in cell lines from high-grade dysplasia (GihTERT) and oesophageal adenocarcinoma (FLO) but not a non-dysplastic Barrett's cell line (QhTERT). Real-time reverse transcription-polymerase chain reaction revealed that NO induced expression of matrix metalloproteinase (MMP)-1, -3, -7, -9 and -10 and tissue inhibitor of metalloproteinase (TIMP)-1, -2 and -3 in these cell lines. Furthermore, ex vivo treatment of Barrett's biopsy samples with NO induced increases in MMP-1 and TIMP-1 expression, suggesting that NO enhances invasion through deregulating MMP and TIMP expression in epithelial cells. In keeping with these findings, microarray analysis and immunohistochemistry performed on biopsy samples showed enhanced expression of MMP-1, -3, -7 and -10 and TIMP-1 in the progression from non-dysplastic BO to adenocarcinoma, although this could not be directly attributed to the effect of NO. Thus, NO may play a role in Barrett's carcinogenesis through deregulating MMP and TIMP expression to enhance invasive potential.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Esophageal Neoplasms/pathology , Free Radical Scavengers/pharmacology , Nitric Oxide/pharmacology , Precancerous Conditions/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Barrett Esophagus/drug therapy , Barrett Esophagus/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Disease Progression , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Gene Expression Profiling , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/drug therapy , Precancerous Conditions/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tumor Cells, Cultured
17.
Proc Natl Acad Sci U S A ; 107(5): 2177-82, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20080664

ABSTRACT

The stromal compartment is increasingly recognized to play a role in cancer. However, its role in the transition from preinvasive to invasive disease is unknown. Most gastrointestinal tumors have clearly defined premalignant stages, and Barrett's esophagus (BE) is an ideal research model. Supervised clustering of gene expression profiles from microdissected stroma identified a gene signature that could distinguish between BE metaplasia, dysplasia, and esophageal adenocarcinoma (EAC). EAC patients overexpressing any of the five genes (TMEPAI, JMY, TSP1, FAPalpha, and BCL6) identified from this stromal signature had a significantly poorer outcome. Gene ontology analysis identified a strong inflammatory component in BE disease progression, and key pathways included cytokine-cytokine receptor interactions and TGF-beta. Increased protein levels of inflammatory-related genes significantly up-regulated in EAC compared with preinvasive stages were confirmed in the stroma of independent samples, and in vitro assays confirmed functional relevance of these genes. Gene set enrichment analysis of external datasets demonstrated that the stromal signature was also relevant in the preinvasive to invasive transition of the stomach, colon, and pancreas. These data implicate inflammatory pathways in the genesis of gastrointestinal tract cancers, which can affect prognosis.


Subject(s)
Digestive System Neoplasms/genetics , Digestive System Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Barrett Esophagus/genetics , Barrett Esophagus/immunology , Barrett Esophagus/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cytokines/genetics , DNA-Binding Proteins/genetics , Digestive System Neoplasms/immunology , Endopeptidases , Esophageal Neoplasms/genetics , Esophageal Neoplasms/immunology , Esophageal Neoplasms/pathology , Gelatinases/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Membrane Proteins/genetics , Metaplasia , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Nuclear Proteins/genetics , Oncogenes , Precancerous Conditions/genetics , Precancerous Conditions/immunology , Precancerous Conditions/pathology , Prognosis , Proto-Oncogene Proteins c-bcl-6 , Receptors, Cytokine/genetics , Serine Endopeptidases/genetics , Stromal Cells/immunology , Stromal Cells/pathology , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...