Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Nature ; 616(7958): 798-805, 2023 04.
Article in English | MEDLINE | ID: mdl-37046089

ABSTRACT

Oncogene amplification on extrachromosomal DNA (ecDNA) drives the evolution of tumours and their resistance to treatment, and is associated with poor outcomes for patients with cancer1-6. At present, it is unclear whether ecDNA is a later manifestation of genomic instability, or whether it can be an early event in the transition from dysplasia to cancer. Here, to better understand the development of ecDNA, we analysed whole-genome sequencing (WGS) data from patients with oesophageal adenocarcinoma (EAC) or Barrett's oesophagus. These data included 206 biopsies in Barrett's oesophagus surveillance and EAC cohorts from Cambridge University. We also analysed WGS and histology data from biopsies that were collected across multiple regions at 2 time points from 80 patients in a case-control study at the Fred Hutchinson Cancer Center. In the Cambridge cohorts, the frequency of ecDNA increased between Barrett's-oesophagus-associated early-stage (24%) and late-stage (43%) EAC, suggesting that ecDNA is formed during cancer progression. In the cohort from the Fred Hutchinson Cancer Center, 33% of patients who developed EAC had at least one oesophageal biopsy with ecDNA before or at the diagnosis of EAC. In biopsies that were collected before cancer diagnosis, higher levels of ecDNA were present in samples from patients who later developed EAC than in samples from those who did not. We found that ecDNAs contained diverse collections of oncogenes and immunomodulatory genes. Furthermore, ecDNAs showed increases in copy number and structural complexity at more advanced stages of disease. Our findings show that ecDNA can develop early in the transition from high-grade dysplasia to cancer, and that ecDNAs progressively form and evolve under positive selection.


Subject(s)
Adenocarcinoma , Barrett Esophagus , Carcinogenesis , DNA , Disease Progression , Early Detection of Cancer , Esophageal Neoplasms , Humans , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Case-Control Studies , DNA/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Carcinogenesis/genetics , Whole Genome Sequencing , Cohort Studies , Biopsy , Oncogenes , Immunomodulation , DNA Copy Number Variations , Gene Amplification , Early Detection of Cancer/methods
2.
Nat Commun ; 13(1): 2300, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35484108

ABSTRACT

While the genomes of normal tissues undergo dynamic changes over time, little is understood about the temporal-spatial dynamics of genomes in premalignant tissues that progress to cancer compared to those that remain cancer-free. Here we use whole genome sequencing to contrast genomic alterations in 427 longitudinal samples from 40 patients with stable Barrett's esophagus compared to 40 Barrett's patients who progressed to esophageal adenocarcinoma (ESAD). We show the same somatic mutational processes are active in Barrett's tissue regardless of outcome, with high levels of mutation, ESAD gene and focal chromosomal alterations, and similar mutational signatures. The critical distinction between stable Barrett's versus those who progress to cancer is acquisition and expansion of TP53-/- cell populations having complex structural variants and high-level amplifications, which are detectable up to six years prior to a cancer diagnosis. These findings reveal the timing of common somatic genome dynamics in stable Barrett's esophagus and define key genomic features specific to progression to esophageal adenocarcinoma, both of which are critical for cancer prevention and early detection strategies.


Subject(s)
Adenocarcinoma , Barrett Esophagus , Esophageal Neoplasms , Adenocarcinoma/pathology , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Disease Progression , Esophageal Neoplasms/pathology , Humans
3.
Evol Appl ; 14(2): 399-415, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33664784

ABSTRACT

Barrett's Esophagus is a neoplastic condition which progresses to esophageal adenocarcinoma in 5% of cases. Key events affecting the outcome likely occur before diagnosis of Barrett's and cannot be directly observed; we use phylogenetic analysis to infer such past events. We performed whole-genome sequencing on 4-6 samples from 40 cancer outcome and 40 noncancer outcome patients with Barrett's Esophagus, and inferred within-patient phylogenies of deconvoluted clonal lineages. Spatially proximate lineages clustered in the phylogenies, but temporally proximate ones did not. Lineages with inferred loss-of-function mutations in both copies of TP53 and CDKN2A showed enhanced spatial spread, whereas lineages with loss-of-function mutations in other frequently mutated loci did not. We propose a two-phase model with expansions of TP53 and CKDN2A mutant lineages during initial growth of the segment, followed by relative stasis. Subsequent to initial expansion, mutations in these loci as well as ARID1A and SMARCA4 may show a local selective advantage but do not expand far: The spatial structure of the Barrett's segment remains stable during surveillance even in patients who go on to cancer. We conclude that the cancer/noncancer outcome is strongly affected by early steps in formation of the Barrett's segment.

4.
Cell ; 183(1): 197-210.e32, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33007263

ABSTRACT

Cancer genomes often harbor hundreds of somatic DNA rearrangement junctions, many of which cannot be easily classified into simple (e.g., deletion) or complex (e.g., chromothripsis) structural variant classes. Applying a novel genome graph computational paradigm to analyze the topology of junction copy number (JCN) across 2,778 tumor whole-genome sequences, we uncovered three novel complex rearrangement phenomena: pyrgo, rigma, and tyfonas. Pyrgo are "towers" of low-JCN duplications associated with early-replicating regions, superenhancers, and breast or ovarian cancers. Rigma comprise "chasms" of low-JCN deletions enriched in late-replicating fragile sites and gastrointestinal carcinomas. Tyfonas are "typhoons" of high-JCN junctions and fold-back inversions associated with expressed protein-coding fusions, breakend hypermutation, and acral, but not cutaneous, melanomas. Clustering of tumors according to genome graph-derived features identified subgroups associated with DNA repair defects and poor prognosis.


Subject(s)
Genomic Structural Variation/genetics , Genomics/methods , Neoplasms/genetics , Chromosome Inversion/genetics , Chromothripsis , DNA Copy Number Variations/genetics , Gene Rearrangement/genetics , Genome, Human/genetics , Humans , Mutation/genetics , Whole Genome Sequencing/methods
5.
Genome Med ; 11(1): 14, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30867038

ABSTRACT

It was highlighted that in the original article [1] the Availability of data and materials section was incorrect.

6.
Genome Med ; 10(1): 17, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29486792

ABSTRACT

BACKGROUND: Use of aspirin and other non-steroidal anti-inflammatory drugs (NSAIDs) has been shown to protect against tetraploidy, aneuploidy, and chromosomal alterations in the metaplastic condition Barrett's esophagus (BE) and to lower the incidence and mortality of esophageal adenocarcinoma (EA). The esophagus is exposed to both intrinsic and extrinsic mutagens resulting from gastric reflux, chronic inflammation, and exposure to environmental carcinogens such as those found in cigarettes. Here we test the hypothesis that NSAID use inhibits accumulation of point mutations/indels during somatic genomic evolution in BE. METHODS: Whole exome sequences were generated from 82 purified epithelial biopsies and paired blood samples from a cross-sectional study of 41 NSAID users and 41 non-users matched by sex, age, smoking, and continuous time using or not using NSAIDs. RESULTS: NSAID use reduced overall frequency of point mutations across the spectrum of mutation types, lowered the frequency of mutations even when adjusted for both TP53 mutation and smoking status, and decreased the prevalence of clones with high variant allele frequency. Never smokers who consistently used NSAIDs had fewer point mutations in signature 17, which is commonly found in EA. NSAID users had, on average, a 50% reduction in functional gene mutations in nine cancer-associated pathways and also had less diversity in pathway mutational burden compared to non-users. CONCLUSIONS: These results indicate NSAID use functions to limit overall mutations on which selection can act and supports a model in which specific mutant cell populations survive or expand better in the absence of NSAIDs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Barrett Esophagus/drug therapy , Barrett Esophagus/genetics , Exome/genetics , Mutation/genetics , DNA Copy Number Variations/genetics , Gene Frequency/genetics , Humans , Loss of Heterozygosity , Mutagenesis/genetics
7.
Nat Commun ; 9(1): 794, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29476056

ABSTRACT

The low risk of progression of Barrett's esophagus (BE) to esophageal adenocarcinoma can lead to over-diagnosis and over-treatment of BE patients. This may be addressed through a better understanding of the dynamics surrounding BE malignant progression. Although genetic diversity has been characterized as a marker of malignant development, it is still unclear how BE arises and develops. Here we uncover the evolutionary dynamics of BE at crypt and biopsy levels in eight individuals, including four patients that experienced malignant progression. We assay eight individual crypts and the remaining epithelium by SNP array for each of 6-11 biopsies over 2 time points per patient (358 samples in total). Our results indicate that most Barrett's segments are clonal, with similar number and inferred rates of alterations observed for crypts and biopsies. Divergence correlates with geographical location, being higher near the gastro-esophageal junction. Relaxed clock analyses show that genomic instability precedes and is enhanced by genome doubling. These results shed light on the clinically relevant evolutionary dynamics of BE.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Esophageal Neoplasms/genetics , Evolution, Molecular , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Biopsy , Disease Progression , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Genomic Instability , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
8.
PLoS One ; 10(7): e0133403, 2015.
Article in English | MEDLINE | ID: mdl-26230607

ABSTRACT

PURPOSE: Goblet cells may represent a potentially successful adaptive response to acid and bile by producing a thick mucous barrier that protects against cancer development in Barrett's esophagus (BE). The aim of this study was to determine the relationship between goblet cells (GC) and risk of progression to adenocarcinoma, and DNA content flow cytometric abnormalities, in BE patients. EXPERIMENTAL DESIGN: Baseline mucosal biopsies (N=2988) from 213 patients, 32 of whom developed cancer during the follow up period, enrolled in a prospective dynamic cohort of BE patients were scored in a blinded fashion, for the total number (#) of GC, mean # of GC/crypt (GC density), # of crypts with ≥ 1 GC, and the proportion of crypts with ≥1 GC, in both dysplastic and non-dysplastic epithelium separately. The relationship between these four GC parameters and DNA content flow cytometric abnormalities and adenocarcinoma outcome was compared, after adjustment for age, gender, and BE segment length. RESULTS: High GC parameters were inversely associated with DNA content flow cytometric abnormalities, such as aneuploidy, ploidy >2.7N, and an elevated 4N fraction > 6%, and with risk of adenocarcinoma. However, a Kaplan-Meier analysis showed that the total # of GC and the total # crypts with ≥1 GC were the only significant GC parameters (p<0.001 and 0.003, respectively). CONCLUSIONS: The results of this study show, for the first time, an inverse relationship between high GC counts and flow cytometric abnormalities and risk of adenocarcinoma in BE. Further studies are needed to determine if GC depleted foci within esophageal columnar mucosa are more prone to neoplastic progression or whether loss of GC occurs secondary to underlying genetic abnormalities.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Esophageal Neoplasms/pathology , Goblet Cells/pathology , Adenocarcinoma/genetics , Aged , Barrett Esophagus/genetics , Biopsy , Case-Control Studies , Cross-Sectional Studies , DNA/genetics , Esophageal Neoplasms/genetics , Female , Flow Cytometry/methods , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Mucous Membrane/pathology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Prospective Studies , Risk
9.
Cancer Prev Res (Phila) ; 8(9): 845-56, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26130253

ABSTRACT

Cancers detected at a late stage are often refractory to treatments and ultimately lethal. Early detection can significantly increase survival probability, but attempts to reduce mortality by early detection have frequently increased overdiagnosis of indolent conditions that do not progress over a lifetime. Study designs that incorporate biomarker trajectories in time and space are needed to distinguish patients who progress to an early cancer from those who follow an indolent course. Esophageal adenocarcinoma is characterized by evolution of punctuated and catastrophic somatic chromosomal alterations and high levels of overall mutations but few recurrently mutated genes aside from TP53. Endoscopic surveillance of Barrett's esophagus for early cancer detection provides an opportunity for assessment of alterations for cancer risk in patients who progress to esophageal adenocarcinoma compared with nonprogressors. We investigated 1,272 longitudinally collected esophageal biopsies in a 248 Barrett's patient case-cohort study with 20,425 person-months of follow-up, including 79 who progressed to early-stage esophageal adenocarcinoma. Cancer progression risk was assessed for total chromosomal alterations, diversity, and chromosomal region-specific alterations measured with single-nucleotide polymorphism arrays in biopsies obtained over esophageal space and time. A model using 29 chromosomal features was developed for cancer risk prediction (area under receiver operator curve, 0.94). The model prediction performance was robust in two independent esophageal adenocarcinoma sets and outperformed TP53 mutation, flow cytometric DNA content, and histopathologic diagnosis of dysplasia. This study offers a strategy to reduce overdiagnosis in Barrett's esophagus and improve early detection of esophageal adenocarcinoma and potentially other cancers characterized by punctuated and catastrophic chromosomal evolution.


Subject(s)
Adenocarcinoma/diagnosis , Barrett Esophagus/diagnosis , Chromosome Aberrations , Esophageal Neoplasms/diagnosis , Risk Assessment/methods , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Biopsy , Cohort Studies , Disease Progression , Endoscopy/methods , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Genome, Human , Humans , Longitudinal Studies , Male , Middle Aged , Mutation , ROC Curve , Stochastic Processes
10.
PLoS One ; 10(6): e0129055, 2015.
Article in English | MEDLINE | ID: mdl-26076489

ABSTRACT

BACKGROUND: The incidence of esophageal adenocarcinoma (EAC) has increased nearly five-fold over the last four decades in the United States. Barrett's esophagus, the replacement of the normal squamous epithelial lining with a mucus-secreting columnar epithelium, is the only known precursor to EAC. Like other parts of the gastrointestinal (GI) tract, the esophagus hosts a variety of bacteria and comparisons among published studies suggest bacterial communities in the stomach and esophagus differ. Chronic infection with Helicobacter pylori in the stomach has been inversely associated with development of EAC, but the mechanisms underlying this association remain unclear. METHODOLOGY: The bacterial composition in the upper GI tract was characterized in a subset of participants (n=12) of the Seattle Barrett's Esophagus Research cohort using broad-range 16S PCR and pyrosequencing of biopsy and brush samples collected from squamous esophagus, Barrett's esophagus, stomach corpus and stomach antrum. Three of the individuals were sampled at two separate time points. Prevalence of H. pylori infection and subsequent development of aneuploidy (n=339) and EAC (n=433) was examined in a larger subset of this cohort. RESULTS/SIGNIFICANCE: Within individuals, bacterial communities of the stomach and esophagus showed overlapping community membership. Despite closer proximity, the stomach antrum and corpus communities were less similar than the antrum and esophageal samples. Re-sampling of study participants revealed similar upper GI community membership in two of three cases. In this Barrett's esophagus cohort, Streptococcus and Prevotella species dominate the upper GI and the ratio of these two species is associated with waist-to-hip ratio and hiatal hernia length, two known EAC risk factors in Barrett's esophagus. H. pylori-positive individuals had a significantly decreased incidence of aneuploidy and a non-significant trend toward lower incidence of EAC.


Subject(s)
Bacteria , Barrett Esophagus/genetics , Barrett Esophagus/microbiology , Genomic Instability , Microbiota , Upper Gastrointestinal Tract/microbiology , Adenocarcinoma/epidemiology , Adenocarcinoma/etiology , Aged , Aged, 80 and over , Bacteria/classification , Bacteria/genetics , Barrett Esophagus/complications , Barrett Esophagus/pathology , Biodiversity , Disease Susceptibility , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/etiology , Female , Gastric Mucosa/metabolism , Humans , Incidence , Male , Metagenome , Microvilli/microbiology , Middle Aged , Mucous Membrane/metabolism , Mucous Membrane/microbiology , Mucous Membrane/pathology , Phylogeny , Quantitative Trait, Heritable , Risk Factors , Stomach/microbiology , Waist-Hip Ratio
11.
Cancer Prev Res (Phila) ; 7(1): 114-27, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24253313

ABSTRACT

All cancers are believed to arise by dynamic, stochastic somatic genomic evolution with genome instability, generation of diversity, and selection of genomic alterations that underlie multistage progression to cancer. Advanced esophageal adenocarcinomas have high levels of somatic copy number alterations. Barrett's esophagus is a risk factor for developing esophageal adenocarcinoma, and somatic chromosomal alterations (SCA) are known to occur in Barrett's esophagus. The vast majority (∼95%) of individuals with Barrett's esophagus do not progress to esophageal adenocarcinoma during their lifetimes, but a small subset develop esophageal adenocarcinoma, many of which arise rapidly even in carefully monitored patients without visible endoscopic abnormalities at the index endoscopy. Using a well-designed, longitudinal case-cohort study, we characterized SCA as assessed by single-nucleotide polymorphism arrays over space and time in 79 "progressors" with Barrett's esophagus as they approach the diagnosis of cancer and 169 "nonprogressors" with Barrett's esophagus who did not progress to esophageal adenocarcinoma over more than 20,425 person-months of follow-up. The genomes of nonprogressors typically had small localized deletions involving fragile sites and 9p loss/copy neutral LOH that generate little genetic diversity and remained relatively stable over prolonged follow-up. As progressors approach the diagnosis of cancer, their genomes developed chromosome instability with initial gains and losses, genomic diversity, and selection of SCAs followed by catastrophic genome doublings. Our results support a model of differential disease dynamics in which nonprogressor genomes largely remain stable over prolonged periods, whereas progressor genomes evolve significantly increased SCA and diversity within four years of esophageal adenocarcinoma diagnosis, suggesting a window of opportunity for early detection.


Subject(s)
Barrett Esophagus/genetics , Chromosome Aberrations , Adenocarcinoma/genetics , Adult , Aged , Biopsy , Case-Control Studies , Chromosomal Instability , Disease Progression , Endoscopy , Esophageal Neoplasms/genetics , Female , Genome, Human , Humans , Longitudinal Studies , Loss of Heterozygosity , Male , Middle Aged , Polymorphism, Single Nucleotide , Time Factors
12.
Obstet Gynecol Int ; 2013: 576842, 2013.
Article in English | MEDLINE | ID: mdl-24282417

ABSTRACT

Identifying molecular markers of endometrial hyperplasia (neoplasia) progression is critical to cancer prevention. To assess RNA and DNA quantity and quality from routinely collected endometrial samples and evaluate the performance of RNA- and DNA-based arrays across endometrial tissue types, we collected fresh frozen (FF) Pipelle, FF curettage, and formalin-fixed paraffin-embedded (FFPE) hysterectomy specimens (benign indications) from eight women. Additionally, neoplastic and uninvolved tissues from 24 FFPE archival hysterectomy specimens with endometrial hyperplasias and carcinomas were assessed. RNA was extracted from 15 of 16 FF and 51 of 51 FFPE samples, with yields >1.2 µ g for 13/15 (87%) FF and 50/51 (98%) FFPE samples. Extracted RNA was of high quality; all samples performed successfully on the Illumina whole-genome cDNA-mediated annealing, selection, extension, and ligation (WG-DASL) array and performance did not vary by tissue type. While DNA quantity from FFPE samples was excellent, quality was not sufficient for successful performance on the Affymetrix SNP Array 6.0. In conclusion, FF Pipelle samples, which are minimally invasive, yielded excellent quantity and quality of RNA for gene expression arrays (similar to FF curettage) and should be considered for use in genomic studies. FFPE-derived DNA should be evaluated on new rapidly evolving sequencing platforms.

13.
PLoS Genet ; 9(6): e1003553, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23785299

ABSTRACT

Cancer is considered an outcome of decades-long clonal evolution fueled by acquisition of somatic genomic abnormalities (SGAs). Non-steroidal anti-inflammatory drugs (NSAIDs) have been shown to reduce cancer risk, including risk of progression from Barrett's esophagus (BE) to esophageal adenocarcinoma (EA). However, the cancer chemopreventive mechanisms of NSAIDs are not fully understood. We hypothesized that NSAIDs modulate clonal evolution by reducing SGA acquisition rate. We evaluated thirteen individuals with BE. Eleven had not used NSAIDs for 6.2±3.5 (mean±standard deviation) years and then began using NSAIDs for 5.6±2.7 years, whereas two had used NSAIDs for 3.3±1.4 years and then discontinued use for 7.9±0.7 years. 161 BE biopsies, collected at 5-8 time points over 6.4-19 years, were analyzed using 1Million-SNP arrays to detect SGAs. Even in the earliest biopsies there were many SGAs (284±246 in 10/13 and 1442±560 in 3/13 individuals) and in most individuals the number of SGAs changed little over time, with both increases and decreases in SGAs detected. The estimated SGA rate was 7.8 per genome per year (95% support interval [SI], 7.1-8.6) off-NSAIDs and 0.6 (95% SI 0.3-1.5) on-NSAIDs. Twelve individuals did not progress to EA. In ten we detected 279±86 SGAs affecting 53±30 Mb of the genome per biopsy per time point and in two we detected 1,463±375 SGAs affecting 180±100 Mb. In one individual who progressed to EA we detected a clone having 2,291±78 SGAs affecting 588±18 Mb of the genome at three time points in the last three of 11.4 years of follow-up. NSAIDs were associated with reduced rate of acquisition of SGAs in eleven of thirteen individuals. Barrett's cells maintained relative equilibrium level of SGAs over time with occasional punctuations by expansion of clones having massive amount of SGAs.


Subject(s)
Adenocarcinoma/genetics , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Barrett Esophagus/genetics , Clonal Evolution/genetics , Genomic Instability/drug effects , Adenocarcinoma/pathology , Aged , Barrett Esophagus/pathology , Biopsy , Clonal Evolution/drug effects , Disease Progression , Female , Humans , Male , Middle Aged , Phylogeny , Polymorphism, Single Nucleotide
14.
PLoS One ; 8(2): e56884, 2013.
Article in English | MEDLINE | ID: mdl-23460817

ABSTRACT

BACKGROUND: Increased glycolysis is a hallmark of cancer metabolism, yet relatively little is known about this phenotype at premalignant stages of progression. Periodic ischemia occurs in the premalignant condition Barrett's esophagus (BE) due to tissue damage from chronic acid-bile reflux and may select for early adaptations to hypoxia, including upregulation of glycolysis. METHODOLOGY/PRINCIPAL FINDINGS: We compared rates of glycolysis and oxidative phosphorylation in four cell lines derived from patients with BE (CP-A, CP-B, CP-C and CP-D) in response to metabolic inhibitors and changes in glucose concentration. We report that cell lines derived from patients with more advanced genetically unstable BE have up to two-fold higher glycolysis compared to a cell line derived from a patient with early genetically stable BE; however, all cell lines preserve active mitochondria. In response to the glycolytic inhibitor 2-deoxyglucose, the most glycolytic cell lines (CP-C and CP-D) had the greatest suppression of extra-cellular acidification, but were able to compensate with upregulation of oxidative phosphorylation. In addition, these cell lines showed the lowest compensatory increases in glycolysis in response to mitochondrial uncoupling by 2,4-dinitrophenol. Finally, these cell lines also upregulated their oxidative phosphorylation in response to glucose via the Crabtree effect, and demonstrate a greater range of modulation of oxygen consumption. CONCLUSIONS/SIGNIFICANCE: Our findings suggest that cells from premalignant Barrett's esophagus tissue may adapt to an ever-changing selective microenvironment through changes in energy metabolic pathways typically associated with cancer cells.


Subject(s)
Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Mitochondria/metabolism , Models, Biological , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , 2,4-Dinitrophenol/pharmacology , Acids/metabolism , Barrett Esophagus/genetics , Cell Line, Tumor , Deoxyglucose/pharmacology , Disease Progression , Energy Metabolism/drug effects , Energy Metabolism/genetics , Extracellular Space/metabolism , Genomic Instability/drug effects , Glycolysis/drug effects , Humans , Mitochondria/drug effects , Neoplasm Staging , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Uncoupling Agents/pharmacology
15.
Mol Cancer Res ; 8(8): 1084-94, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20647332

ABSTRACT

Barrett's esophagus (BE) is a premalignant intermediate to esophageal adenocarcinoma, which develops in the context of chronic inflammation and exposure to bile and acid. We asked whether there might be common genomic alterations that could be identified as potential clinical biomarker(s) for BE by whole genome profiling. We detected copy number alterations and/or loss of heterozygosity at 56 fragile sites in 20 patients with premalignant BE. Chromosomal fragile sites are particularly sensitive to DNA breaks and are frequent sites of rearrangement or loss in many human cancers. Seventy-eight percent of all genomic alterations detected by array-CGH were associated with fragile sites. Copy number losses in early BE were observed at particularly high frequency at FRA3B (81%), FRA9A/C (71.4%), FRA5E (52.4%), and FRA 4D (52.4%), and at lower frequencies in other fragile sites, including FRA1K (42.9%), FRAXC (42.9%), FRA 12B (33.3%), and FRA16D (33.3%). Due to the consistency of the region of copy number loss, we were able to verify these results by quantitative PCR, which detected the loss of FRA3B and FRA16D, in 83% and 40% of early molecular stage BE patients, respectively. Loss of heterozygosity in these cases was confirmed through pyrosequencing at FRA3B and FRA16D (75% and 70%, respectively). Deletion and genomic instability at FRA3B and other fragile sites could thus be a biomarker of genetic damage in BE patients and a potential biomarker of cancer risk.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Chromosome Fragile Sites/genetics , Esophageal Neoplasms/genetics , Genomic Instability , Loss of Heterozygosity , Neoplasm Proteins/genetics , Adenocarcinoma/pathology , Barrett Esophagus/pathology , Chromosome Fragility , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 9/genetics , Comparative Genomic Hybridization , Esophageal Neoplasms/pathology , Gene Dosage , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Clin Cancer Res ; 15(10): 3305-14, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19417022

ABSTRACT

PURPOSE: Chromosomal instability, as assessed by many techniques, including DNA content aneuploidy, loss of heterozygosity, and comparative genomic hybridization, has consistently been reported to be common in cancer and rare in normal tissues. Recently, a panel of chromosome instability biomarkers, including loss of heterozygosity and DNA content, has been reported to identify patients at high and low risk of progression from Barrett's esophagus (BE) to esophageal adenocarcinoma (EA), but required multiple platforms for implementation. Although chromosomal instability involving amplifications and deletions of chromosome regions have been observed in nearly all cancers, copy number alterations (CNA) in premalignant tissues have not been well characterized or evaluated in cohort studies as biomarkers of cancer risk. EXPERIMENTAL DESIGN: We examined CNAs in 98 patients having either BE or EA using Bacterial Artificial Chromosome (BAC) array comparative genomic hybridization to characterize CNAs at different stages of progression ranging from early BE to advanced EA. RESULTS: CNAs were rare in early stages (less than high-grade dysplasia) but were progressively more frequent and larger in later stages (high-grade dysplasia and EA), including high-level amplifications. The number of CNAs correlated highly with DNA content aneuploidy. Patients whose biopsies contained CNAs involving >70 Mbp were at increased risk of progression to DNA content abnormalities or EA (hazards ratio, 4.9; 95% confidence interval, 1.6-14.8; P = 0.0047), and the risk increased as more of the genome was affected. CONCLUSIONS: Genome-wide analysis of CNAs provides a common platform for the evaluation of chromosome instability for cancer risk assessment as well as for the identification of common regions of alteration that can be further studied for biomarker discovery.


Subject(s)
Barrett Esophagus/genetics , Chromosomal Instability , Esophageal Neoplasms/genetics , Gene Dosage , Aged , Barrett Esophagus/pathology , Chromosome Aberrations , Comparative Genomic Hybridization/methods , DNA, Neoplasm/metabolism , Disease Progression , Esophageal Neoplasms/pathology , Flow Cytometry , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Loss of Heterozygosity , Middle Aged , Neoplasm Staging
17.
Am J Surg Pathol ; 33(6): 886-93, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19194279

ABSTRACT

Rarely, dysplasia in Barrett's esophagus (BE) is composed of crypts lined by cuboidal-shaped cells that contain a centrally located nucleus, markedly increased nuclear/cytoplasmic ratio, but without nuclear stratification characteristic of conventional "adenomatous" dysplasia. The aim of this study was to evaluate the clinical and pathologic features, natural history, and DNA content flow cytometric abnormalities of BE patients with non-adenomatous dysplasia (NAD) in a cohort of BE patients enrolled in a prospective surveillance program. Eighteen patients with NAD identified over a 6 year period, in a cohort of 270 consecutive patients with BE and without esophageal adenocarcinoma (EA) at baseline, were evaluated for clinical and pathologic features, including association with conventional adenomatous dysplasia and EA, DNA content flow cytometric abnormalities (tetraploidy and aneuploidy) and outcome, over a mean follow-up period of 4.1 years. The findings in the 18 study patients were compared to those in the 252 remaining (control) patients without NAD. Control patients included 228 with metaplasia/indefinite for dysplasia, and 24 with conventional adenomatous dysplasia (13 low-grade, 11 high-grade). The prevalence rate of NAD in our BE cohort was 6.7% Of the 18 study patients, there were 17 were males and 1 female of mean age 66.7 years. The mean length of BE was 3.9 cm NAD foci were associated with goblet or non-goblet epithelium in 62% and 38% of cases, respectively. Ninety-four percent of patients with NAD (17/18) also had conventional adenomatous dysplasia (four with low-grade, 13 with high-grade) elsewhere in the esophagus at the same endoscopic procedure as the one that detected NAD. Patients with NAD had a significantly shorter length of BE compared to control patients with conventional adenomatous dysplasia (N=24) (p=0.03). Patients with NAD also showed a significantly higher rate of DNA content flow cytometric abnormalities compared to the entire cohort of control patients (38% vs. 11%, p=0.05). However, no significant differences regarding either flow cytometric abnormalities or progression to EA were found when the NAD patients were compared only to the 24 controls with conventional adenomatous dysplasia. NAD is a high grade histologic variant of intraepithelial neoplasia that is episodic in nature, and shows a high association with conventional adenomatous high-grade dysplasia.


Subject(s)
Barrett Esophagus/genetics , Barrett Esophagus/pathology , Adenocarcinoma/epidemiology , Adenocarcinoma/pathology , Aged , Disease Progression , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/pathology , Female , Flow Cytometry , Humans , Male , Metaplasia/epidemiology , Metaplasia/pathology , Prevalence , Risk Factors
18.
PLoS One ; 3(11): e3809, 2008.
Article in English | MEDLINE | ID: mdl-19043591

ABSTRACT

BACKGROUND: Mutation, promoter hypermethylation and loss of heterozygosity involving the tumor suppressor gene p16 (CDKN2a/INK4a) have been detected in a wide variety of human cancers, but much less is known concerning the frequency and spectrum of p16 mutations in premalignant conditions. METHODS AND FINDINGS: We have determined the p16 mutation spectrum for a cohort of 304 patients with Barrett's esophagus, a premalignant condition that predisposes to the development of esophageal adenocarcinoma. Forty seven mutations were detected by sequencing of p16 exon 2 in 44 BE patients (14.5%) with a mutation spectrum consistent with that caused by oxidative damage and chronic inflammation. The percentage of patients with p16 mutations increased with increasing histologic grade. In addition, samples from 3 out of 19 patients (15.8%) who underwent esophagectomy were found to have mutations. CONCLUSIONS: The results of this study suggest the environment of the esophagus in BE patients can both generate and select for clones with p16 mutations.


Subject(s)
Barrett Esophagus/genetics , Genes, p16 , Mutation , Precancerous Conditions/genetics , Adult , Aged , Aged, 80 and over , Clone Cells , Cohort Studies , DNA Mutational Analysis , Esophagus/pathology , Exons , Female , Humans , Male , Middle Aged , Young Adult
19.
Clin Cancer Res ; 14(21): 6988-95, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18980994

ABSTRACT

PURPOSE: Elevated cellular proliferation and cell cycle abnormalities, which have been associated with premalignant lesions, may be caused by inactivation of tumor suppressor genes. We measured proliferative and cell cycle fractions of biopsies from a cohort of patients with Barrett's esophagus to better understand the role of proliferation in early neoplastic progression and the association between cell cycle dysregulation and tumor suppressor gene inactivation. EXPERIMENTAL DESIGN: Cell proliferative fractions (determined by Ki67/DNA multiparameter flow cytometry) and cell cycle fractions (DNA content flow cytometry) were measured in 853 diploid biopsies from 362 patients with Barrett's esophagus. The inactivation status of CDKN2A and TP53 was assessed in a subset of these biopsies in a cross-sectional study. A prospective study followed 276 of the patients without detectable aneuploidy for an average of 6.3 years with esophageal adenocarcinoma as an end point. RESULTS: Diploid S and 4N (G(2)/tetraploid) fractions were significantly higher in biopsies with TP53 mutation and loss of heterozygosity. CDKN2A inactivation was not associated with higher Ki67-positive, diploid S, G(1), or 4N fractions. High Ki67-positive and G(1)-phase fractions were not associated with the future development of esophageal adenocarcinoma (P=0.13 and P=0.15, respectively), whereas high diploid S-phase and 4N fractions were (P=0.03 and P<0.0001, respectively). CONCLUSIONS: High Ki67-positive proliferative fractions were not associated with inactivation of CDKN2A and TP53 or future development of cancer in our cohort of patients with Barrett's esophagus. Biallelic inactivation of TP53 was associated with elevated 4N fractions, which have been associated with the future development of esophageal adenocarcinoma.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/complications , Barrett Esophagus/pathology , Cell Cycle , Cell Division , Esophageal Neoplasms/pathology , Adenocarcinoma/diagnosis , Adult , Aged , Cohort Studies , Esophageal Neoplasms/etiology , Female , Genes, p16 , Genes, p53 , Humans , Ki-67 Antigen/metabolism , Longitudinal Studies , Loss of Heterozygosity , Male , Middle Aged , Mutation , Prospective Studies
20.
Nutr Cancer ; 60(1): 39-48, 2008.
Article in English | MEDLINE | ID: mdl-18444134

ABSTRACT

The incidence of esophageal adenocarcinoma (EA) and its precursor condition, Barrett's esophagus, has risen rapidly in the United States for reasons that are not fully understood. Therefore, we evaluated the association between use of supplemental vitamins and minerals and risk of neoplastic progression of Barrett's esophagus and EA. The Seattle Barrett's Esophagus Program is a prospective study based on 339 men and women with histologically confirmed Barrett's esophagus. Participants underwent baseline and periodic follow-up exams, which included endoscopy and self-administered questionnaires on diet, supplement use, and lifestyle characteristics. Use of multivitamins and 4 individual supplements was calculated using time-weighted averages of reported use over the observational period. Cox proportional-hazards models were used to calculate hazard ratios (HR) for each endpoint: EA, tetraploidy, and aneuploidy. During a mean follow-up of 5 yr, there were 37 cases of EA, 42 cases of tetraploidy, and 34 cases of aneuploidy. After controlling for multiple covariates including diet, nonsteroidal anti-inflammatory drug use, obesity, and smoking, participants who took 1 or more multivitamin pills/day had a significantly decreased risk of tetraploidy [HR = 0.19; 95% confidence interval (CI) = 0.08-0.47) and EA (HR = 0.38; 95% CI = 0.15-0.99] compared to those not taking multivitamins. Significant inverse associations were also observed between risk of EA and supplemental vitamin C (> or = 250 mg vs. none: HR = 0.25; 95% CI = 0.11-0.58) and vitamin E (> or = 180 mg vs. none: HR = 0.25; 95% CI = 0.10-0.60). In this cohort study, use of multivitamins and single antioxidant supplements was associated with a significantly reduced risk of EA and markers of neoplastic progression among individuals with Barrett's esophagus.


Subject(s)
Adenocarcinoma/epidemiology , Barrett Esophagus/epidemiology , Chemoprevention/methods , Esophageal Neoplasms/epidemiology , Vitamins/administration & dosage , Adenocarcinoma/pathology , Adenocarcinoma/prevention & control , Adult , Aged , Aneuploidy , Ascorbic Acid/administration & dosage , Barrett Esophagus/complications , Barrett Esophagus/pathology , Cell Transformation, Neoplastic/drug effects , Cohort Studies , Dietary Supplements , Disease Progression , Drug Therapy, Combination , Endoscopy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/prevention & control , Female , Flow Cytometry , Follow-Up Studies , Humans , Male , Middle Aged , Proportional Hazards Models , Prospective Studies , Risk Factors , Vitamin E/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...