Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
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
2.
Nat Genet ; 38(4): 468-73, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16565718

ABSTRACT

Neoplasms are thought to progress to cancer through genetic instability generating cellular diversity and clonal expansions driven by selection for mutations in cancer genes. Despite advances in the study of molecular biology of cancer genes, relatively little is known about evolutionary mechanisms that drive neoplastic progression. It is unknown, for example, which may be more predictive of future progression of a neoplasm: genetic homogenization of the neoplasm, possibly caused by a clonal expansion, or the accumulation of clonal diversity. Here, in a prospective study, we show that clonal diversity measures adapted from ecology and evolution can predict progression to adenocarcinoma in the premalignant condition known as Barrett's esophagus, even when controlling for established genetic risk factors, including lesions in TP53 (p53; ref. 6) and ploidy abnormalities. Progression to cancer through accumulation of clonal diversity, on which natural selection acts, may be a fundamental principle of neoplasia with important clinical implications.


Subject(s)
Adenocarcinoma/genetics , Esophageal Neoplasms/genetics , Adenocarcinoma/pathology , Disease Progression , Esophageal Neoplasms/pathology , Genes, p16 , Genes, p53 , Humans , In Situ Hybridization, Fluorescence , Loss of Heterozygosity
3.
Carcinogenesis ; 35(12): 2740-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25280564

ABSTRACT

Incidence of esophageal adenocarcinoma (EA) in Western countries has increased markedly in recent decades. Although several risk factors have been identified for EA and its precursor, Barrett's esophagus (BE), including reflux, Caucasian race, male gender, obesity, and smoking, less is known about the role of inherited genetic variation. Frequent somatic mutations in the tumor suppressor genes CDKN2A and TP53 were recently reported in EA tumors, while somatic alterations at 9p (CDKN2A) and 17p (TP53) have been implicated as predictors of progression from BE to EA. Motivated by these findings, we used data from a genome-wide association study of 2515 EA cases and 3207 controls to analyze 37 germline single nucleotide polymorphisms at the CDKN2A and TP53 loci. Three CDKN2A polymorphisms were nominally associated (P < 0.05) with reduced risk of EA: rs2518720 C>T [intronic, odds ratio 0.90, P = 0.0121, q = 0.3059], rs3088440 G>A (3'UTR, odds ratio 0.84, P = 0.0186, q = 0.3059), and rs4074785 C>T (intronic, odds ratio 0.85, P = 0.0248, q = 0.3059). None of the TP53 single nucleotide polymorphisms reached nominal significance. Two of the CDKN2A variants identified were also associated with reduced risk of progression from BE to EA, when assessed in a prospective cohort of 408 BE patients: rs2518720 (hazard ratio 0.57, P = 0.0095, q = 0.0285) and rs3088440 (hazard ratio 0.34, P = 0.0368, q = 0.0552). In vitro functional studies of rs3088440, a single nucleotide polymorphism located in the seed sequence of a predicted miR-663b binding site, suggested a mechanism whereby the G>A substitution may attenuate miR-663b-mediated repression of the CDKN2A transcript. This study provides the first evidence that germline variation at the CDKN2A locus may influence EA susceptibility.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Esophageal Neoplasms/genetics , Genome-Wide Association Study , Polymorphism, Single Nucleotide/genetics , Tumor Suppressor Protein p53/genetics , Adenocarcinoma/pathology , Aged , Barrett Esophagus/pathology , Case-Control Studies , Disease Progression , Esophageal Neoplasms/pathology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Risk Factors
4.
Cancer Causes Control ; 25(8): 969-76, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24839050

ABSTRACT

PURPOSE: Systemic measures of chronic inflammation, often based on a single blood draw, are frequently used to study the associations between inflammation and chronic diseases such as cancer. However, more information is needed on the measurement error in these markers due to laboratory error, within-person variation over time, and long-term storage. METHODS: We investigated the intraindividual variability of inflammation markers C-reactive protein (CRP), interleukin-6 (IL-6), and soluble tumor necrosis factor receptors I and II (sTNFRI and II) in a subsample of the Seattle Barrett's esophagus study cohort. Two fasting blood samples were collected between 1995 and 2009 from 360 participants on average 1.8 years apart. CRP, IL-6, and sTNF receptor levels were measured by immunonephelometry, ELISA, and multiplex assays, respectively. Intra- and inter-batch coefficients of variation (CV) were estimated using blinded pooled samples within each batch. Intraclass correlations (ICCs) were computed using random effects ANOVA. RESULTS: Intra- and inter-batch CVs for the pooled plasma aliquots were low (2.4-8.9 %), suggesting little laboratory variability. Reliability over time was excellent for sTNF receptors (ICCsTNF-RI = 0.89, ICCsTNF-RII = 0.85) and fair-to-good for CRP and IL-6 (ICCCRP = 0.55, ICCIL-6 = 0.57). For samples stored for over 13 years, the ICCs for CRP and IL-6 were decreased but those for sTNF receptors were unaffected. CONCLUSION: sTNF receptor levels are more stable within person over time than CRP or IL-6. Long-term storage of samples appears to increase the variability of CRP and IL-6 measures, while the reliability of soluble TNF receptor measures was not affected by storage time.


Subject(s)
Inflammation/blood , Aged , Analysis of Variance , Biomarkers/blood , C-Reactive Protein/metabolism , Cohort Studies , Female , Humans , Interleukin-6/blood , Male , Middle Aged , Prospective Studies , Receptors, Tumor Necrosis Factor, Type I/blood , Receptors, Tumor Necrosis Factor, Type II/blood , Specimen Handling , Time Factors
5.
BMC Med Inform Decis Mak ; 14: 15, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24602132

ABSTRACT

BACKGROUND: With the rapid development of "-omic" technologies, an increasing number of purported biomarkers have been identified for cancer and other diseases. The process of identifying those that are most promising and validating them for use at the population level for prevention and early detection is a critical next step in achieving significant health benefits. METHODS: In this paper, we propose that in order to effectively translate biomarkers for practical clinical use, it is important to distinguish and quantify the differences between the use of biomarkers and other risk factors to identify preventive interventions versus their use in disease risk prediction and early detection. We developed mathematical models for quantitatively evaluating risk and benefit in use of biomarkers for disease prevention or early detection. Simple numerical examples were used to demonstrate the potential applications of the models for various types of data. RESULTS: We propose an index which takes into account potential adverse consequences of biomarker-driven interventions - the 'naïve' ratio of population benefit (RPB) - to facilitate evaluating the potential impact of biomarkers on cancer prevention and personalized medicine. The index RPB is developed for both binary and continuous biomarkers/risk factors. Examples with computational analyses are presented in the paper to contrast the differences in using biomarkers/risk factors for prevention and early detection. CONCLUSIONS: Integrating epidemiologic knowledge into clinical decision making is a key step to translate new biomarkers/risk factors into practical use to achieve health benefits. The RPB proposed in this paper considers the absolute risk of a disease in intervention, and takes into account the risk-benefit effects simultaneously for a marker/exposure at the population level. The RPB illustrates a unique approach to quantitatively assess the risk and potential benefits of using a biomarker/risk factor for intervention in both early detection and prevention.


Subject(s)
Biomarkers , Early Diagnosis , Epidemiologic Factors , Models, Theoretical , Primary Prevention , Biomarkers/analysis , Humans , Risk Assessment , Risk Factors , Sensitivity and Specificity
6.
Clin Gastroenterol Hepatol ; 11(8): 934-43, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23466711

ABSTRACT

BACKGROUND & AIMS: Individuals with Barrett's esophagus (BE) have an increased risk of developing esophageal adenocarcinoma (EA). Obesity contributes to the development of BE and its progression to cancer. We investigated the roles of obesity-induced hyperinsulinemia and dysregulation of adipokines in these processes. METHODS: We measured fasting levels of glucose, insulin, leptin, and adiponectin in 392 patients enrolled in the Seattle Barrett's Esophagus Study. We calculated homeostatic model assessment scores (a measure of insulin sensitivity) and identified subjects with metabolic syndrome. We evaluated the association between these measures and the risk of EA using Cox regression models adjusted for known risk factors. RESULTS: Increasing homeostatic model assessment scores were associated with an increasing risk for EA; the strongest association was observed within the first 3 years after participants entered the study (hazard ratio [HR], 2.45; 95% confidence interval [CI], 1.43-4.1; P trend = .001). Leptin level also was associated significantly with an increased risk of EA within 3 years (HR, 2.51; 95% CI, 1.09-5.81; P trend = .03) and 6 years (HR, 2.07; 95% CI, 1.01-4.26; P trend = .048) of baseline. The level of high-molecular-weight adiponectin had a nonlinear inverse association with risk of EA; the strongest associations were observed in the second tertile (HR, 0.34; 95% CI, 0.14-0.82). Metabolic syndrome was not associated with risk of EA. CONCLUSIONS: Among patients with BE, increased levels of leptin and insulin resistance are associated with increased risk for EA, whereas increased levels of high-molecular-weight adiponectin is associated inversely with EA. These biomarkers might be used to determine cancer risk among patients with BE.


Subject(s)
Adenocarcinoma/epidemiology , Barrett Esophagus/complications , Biomarkers/blood , Esophageal Neoplasms/epidemiology , Obesity/complications , Adiponectin/blood , Adult , Aged , Aged, 80 and over , Blood Glucose , Female , Humans , Insulin/blood , Leptin/blood , Male , Metabolic Syndrome/complications , Middle Aged , Risk Assessment
7.
PLoS Comput Biol ; 7(2): e1001087, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21390279

ABSTRACT

Aside from primary prevention, early detection remains the most effective way to decrease mortality associated with the majority of solid cancers. Previous cancer screening models are largely based on classification of at-risk populations into three conceptually defined groups (normal, cancer without symptoms, and cancer with symptoms). Unfortunately, this approach has achieved limited successes in reducing cancer mortality. With advances in molecular biology and genomic technologies, many candidate somatic genetic and epigenetic "biomarkers" have been identified as potential predictors of cancer risk. However, none have yet been validated as robust predictors of progression to cancer or shown to reduce cancer mortality. In this Perspective, we first define the necessary and sufficient conditions for precise prediction of future cancer development and early cancer detection within a simple physical model framework. We then evaluate cancer risk prediction and early detection from a dynamic clonal evolution point of view, examining the implications of dynamic clonal evolution of biomarkers and the application of clonal evolution for cancer risk management in clinical practice. Finally, we propose a framework to guide future collaborative research between mathematical modelers and biomarker researchers to design studies to investigate and model dynamic clonal evolution. This approach will allow optimization of available resources for cancer control and intervention timing based on molecular biomarkers in predicting cancer among various risk subsets that dynamically evolve over time.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Disease Management , Humans , Neoplasms/diagnosis , Predictive Value of Tests , Risk Factors , Stochastic Processes , Systems Biology
8.
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
9.
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
10.
PLoS Med ; 4(2): e67, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17326708

ABSTRACT

BACKGROUND: Somatic genetic CDKN2A, TP53, and DNA content abnormalities are common in many human cancers and their precursors, including esophageal adenocarcinoma (EA) and Barrett's esophagus (BE), conditions for which aspirin and other nonsteroidal anti-inflammatory drugs (NSAIDs) have been proposed as possible chemopreventive agents; however, little is known about the ability of a biomarker panel to predict progression to cancer nor how NSAID use may modulate progression. We aimed to evaluate somatic genetic abnormalities with NSAIDs as predictors of EA in a prospective cohort study of patients with BE. METHODS AND FINDINGS: Esophageal biopsies from 243 patients with BE were evaluated at baseline for TP53 and CDKN2A (p16) alterations, tetraploidy, and aneuploidy using sequencing; loss of heterozygosity (LOH); methylation-specific PCR; and flow cytometry. At 10 y, all abnormalities, except CDKN2A mutation and methylation, contributed to EA risk significantly by univariate analysis, ranging from 17p LOH (relative risk [RR] = 10.6; 95% confidence interval [CI] 5.2-21.3, p < 0.001) to 9p LOH (RR = 2.6; 95% CI 1.1-6.0, p = 0.03). A panel of abnormalities including 17p LOH, DNA content tetraploidy and aneuploidy, and 9p LOH was the best predictor of EA (RR = 38.7; 95% CI 10.8-138.5, p < 0.001). Patients with no baseline abnormality had a 12% 10-y cumulative EA incidence, whereas patients with 17p LOH, DNA content abnormalities, and 9p LOH had at least a 79.1% 10-y EA incidence. In patients with zero, one, two, or three baseline panel abnormalities, there was a significant trend toward EA risk reduction among NSAID users compared to nonusers (p = 0.01). The strongest protective effect was seen in participants with multiple genetic abnormalities, with NSAID nonusers having an observed 10-y EA risk of 79%, compared to 30% for NSAID users (p < 0.001). CONCLUSIONS: A combination of 17p LOH, 9p LOH, and DNA content abnormalities provided better EA risk prediction than any single TP53, CDKN2A, or DNA content lesion alone. NSAIDs are associated with reduced EA risk, especially in patients with multiple high-risk molecular abnormalities.


Subject(s)
Adenocarcinoma/genetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA/genetics , Esophageal Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adenocarcinoma/epidemiology , Adult , Aged , Aged, 80 and over , Aneuploidy , Barrett Esophagus/pathology , Biopsy , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 9 , DNA Methylation , Esophageal Neoplasms/epidemiology , Esophagoscopy , Esophagus/pathology , Female , Humans , Incidence , Longitudinal Studies , Loss of Heterozygosity , Male , Middle Aged , Mutation , Prospective Studies , Risk Assessment
11.
Cancer Epidemiol Biomarkers Prev ; 16(11): 2387-95, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18006928

ABSTRACT

BACKGROUND: Insulin-like growth factor-I (IGF-I) is a potent mitogen. IGF-I and its main binding protein, IGF binding protein-3 (IGFBP-3), and their polymorphisms have been investigated in relation to risk of many cancers, but not esophageal adenocarcinoma. MATERIALS AND METHODS: We used data and specimens from a longitudinal study of persons with Barrett's esophagus (n=344; median, 5.4 years follow up) to determine whether baseline serum concentrations of IGF-I and IGFBP-3 and associated polymorphisms were related to the risk of developing esophageal adenocarcinoma or flow cytometric abnormalities. RESULTS: Overall, circulating concentrations of IGF-I and IGBP-3 were not associated with risk of esophageal adenocarcinoma or flow cytometric abnormalities, with the exception of an approximate tripling of risk of aneuploidy among participants with higher IGFBP-3 levels [above median; adjusted hazard ratio (HR) comparing subjects with levels lower than median versus higher of equal to median, 2.7; 95% confidence interval (95% CI), 1.2-6.0; P=0.01]. Genotypic analyses revealed that persons with the IGF-I [cytosine-adenine (CA)](19) or the IGFBP-3 A-202C C allele were associated with lower circulating concentrations of IGF-I (P(trend)=0.01) and IGFBP-3 (P(trend)=0.002), respectively. Persons with two copies of the IGF-I receptors 2-bp deletion allele had a nonsignificant 2-fold increased risk of tetraploidy (HR, 2.3; 95% CI, 0.9-5.9; P(trend)=0.11). After adjustment for IGFBP-3 levels, participants carrying two IGFBP-3 C alleles had a significantly higher risk of developing aneuploidy (HR, 3.8; 95% CI, 1.0-14.0; P(trend)=0.04) than carriers of A alleles; whereas no associations were observed between the outcomes studied and the IGF-I receptors AGG trinucleotide repeat polymorphism at position 97. CONCLUSION: Our findings, although based on a relatively small number of outcomes and subject to several limitations, indicate a potential role of the complex IGF system in neoplastic progression among persons with Barrett's esophagus.


Subject(s)
Barrett Esophagus/genetics , Barrett Esophagus/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor I/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Barrett Esophagus/metabolism , Disease Progression , Esophageal Neoplasms/metabolism , Female , Genotype , Humans , Insulin-Like Growth Factor Binding Protein 3 , Insulin-Like Growth Factor Binding Proteins/blood , Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Longitudinal Studies , Male , Middle Aged , Ploidies , Polymorphism, Genetic , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism
12.
Cancer Epidemiol Biomarkers Prev ; 16(12): 2649-55, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18086770

ABSTRACT

PURPOSE: Leukocyte telomere length has gained attention as a marker of oxidative damage and age-related diseases, including cancer. We hypothesize that leukocyte telomere length might be able to predict future risk of cancer and examined this in a cohort of patients with Barrett's esophagus, who are at increased risk of esophageal adenocarcinoma and thus were enrolled in a long-term cancer surveillance program. PATIENTS AND METHODS: In this prospective study, telomere length was measured by quantitative PCR in baseline blood samples in a cohort of 300 patients with Barrett's esophagus followed for a mean of 5.8 years. Leukocyte telomere length hazard ratios (HR) for risk of esophageal adenocarcinoma were calculated using multivariate Cox models. RESULTS: Shorter telomeres were associated with increased esophageal adenocarcinoma risk (age-adjusted HR between top and bottom quartiles of telomere length, 3.45; 95% confidence interval, 1.35-8.78; P = 0.009). This association was still significant when individually or simultaneously adjusted for age, gender, nonsteroidal anti-inflammatory drug (NSAID) use, cigarette smoking, and waist-to-hip ratio (HR, 4.18; 95% confidence interval, 1.60-10.94; P = 0.004). The relationship between telomere length and cancer risk was particularly strong among NSAID nonusers, ever smokers, and patients with low waist-to-hip ratio. CONCLUSION: Leukocyte telomere length predicts risk of esophageal adenocarcinoma in patients with Barrett's esophagus independently of smoking, obesity, and NSAID use. These results show the ability of leukocyte telomere length to predict the risk of future cancer and suggest that it might also have predictive value in other cancers arising in a setting of chronic inflammation.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Biomarkers, Tumor/analysis , Esophageal Neoplasms/pathology , Leukocytes/physiology , Telomere/metabolism , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Humans , Polymerase Chain Reaction , Precancerous Conditions/pathology , Risk Factors , Smoking/adverse effects , Waist-Hip Ratio
13.
Clin Cancer Res ; 12(6): 1701-6, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16551852

ABSTRACT

PURPOSE: Neosquamous epithelium (NSE) can arise within Barrett's esophagus as a consequence of medical or surgical acid reduction therapy, as well as after endoscopic ablation. Morphologic studies have suggested that NSE can develop from adjacent squamous epithelium, submucosal gland ducts, or multipotent progenitor cell(s) that can give rise to either squamous or Barrett's epithelium, depending on the luminal environment. The cells responsible for Barrett's epithelium self-renewal are frequently mutated during neoplastic progression. If NSE arises from the same cells that self-renew the Barrett's epithelium, the two tissues should be clonally related and share genetic alterations; if NSE does not originate in the self-renewing Barrett's, NSE and Barrett's esophagus should be genetically independent. EXPERIMENTAL DESIGN: We isolated islands of NSE and the surrounding Barrett's epithelium from 20 patients by microdissection and evaluated each tissue for genetic alterations in exon 2 of CDKN2A or exons 5 to 9 of the TP53 gene. Nine patients had p16 mutations and 11 had TP53 mutations within the Barrett's epithelium. RESULTS: In 1 of 20 patients, a focus of NSE had a 146 bp deletion in p16 identical to that found in surrounding Barrett's epithelium. The NSE in the remaining 19 patients was wild-type for p16 or TP53. CONCLUSION: Our mutational data support the hypothesis that, in most circumstances, NSE originates in cells different from those responsible for self-renewal of Barrett's epithelium. However, in one case, NSE and Barrett's epithelium seem to have arisen from a progenitor cell that was capable of differentiating into either intestinal metaplasia or NSE.


Subject(s)
Barrett Esophagus/pathology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Epithelium/pathology , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Aged , Barrett Esophagus/genetics , Base Sequence , DNA Mutational Analysis , Epithelium/metabolism , Female , Humans , Loss of Heterozygosity , Male , Microdissection
14.
Clin Cancer Res ; 12(12): 3661-97, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778094

ABSTRACT

This article reviews progress in chemopreventive drug development, especially data and concepts that are new since the 2002 AACR report on treatment and prevention of intraepithelial neoplasia. Molecular biomarker expressions involved in mechanisms of carcinogenesis and genetic progression models of intraepithelial neoplasia are discussed and analyzed for how they can inform mechanism-based, molecularly targeted drug development as well as risk stratification, cohort selection, and end-point selection for clinical trials. We outline the concept of augmenting the risk, mechanistic, and disease data from histopathologic intraepithelial neoplasia assessments with molecular biomarker data. Updates of work in 10 clinical target organ sites include new data on molecular progression, significant completed trials, new agents of interest, and promising directions for future clinical studies. This overview concludes with strategies for accelerating chemopreventive drug development, such as integrating the best science into chemopreventive strategies and regulatory policy, providing incentives for industry to accelerate preventive drugs, fostering multisector cooperation in sharing clinical samples and data, and creating public-private partnerships to foster new regulatory policies and public education.


Subject(s)
Biomarkers, Tumor/analysis , Neoplasms, Glandular and Epithelial/prevention & control , Breast Neoplasms/prevention & control , Chemoprevention , Colorectal Neoplasms/prevention & control , Disease Progression , Female , Humans , Infections , Inflammation , Male , Monitoring, Physiologic , Signal Transduction
15.
Cancer Epidemiol Biomarkers Prev ; 15(8): 1451-7, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16896031

ABSTRACT

Barrett's esophagus is a useful model for the study of carcinogenesis, as the metaplastic columnar epithelium that replaces squamous esophageal epithelium is at elevated risk for development of adenocarcinoma. We examined telomere length and chromosomal instability (CIN) in Barrett's esophagus biopsies using fluorescence in situ hybridization. To study CIN, we selected centromere and locus-specific arm probes to chromosomes 17/17p (p53), 11/11q (cyclin D1), and 9/9p (p16 INK4A), loci reported to be involved in early stages of Barrett's esophagus neoplasia. Telomere shortening was observed in Barrett's esophagus epithelium at all histologic grades, whereas CIN was highest in biopsies with dysplastic changes; there was, however, considerable heterogeneity between patients in each variable. Alterations on chromosome 17 were strongly correlated with telomere length (r = 0.55; P < 0.0001) and loss of the 17p arm signal was the most common event. CIN on chromosome 11 was also associated with telomere shortening (r =0.3; P = 0.05), although 11q arm gains were most common. On chromosome 9p, arm losses were the most common finding, but chromosome 9 CIN was not strongly correlated with telomere length. We conclude that CIN is related to telomere shortening in Barrett's esophagus but varies by chromosome. Whether instability is manifested as loss or gain seems to be influenced by the chromosomal loci involved. Because telomere shortening and CIN are early events in Barrett's esophagus neoplastic progression and are highly variable among patients, it will be important to determine whether they identify a subset of patients that is at risk for more rapid neoplastic evolution.


Subject(s)
Barrett Esophagus/genetics , Chromosomal Instability , Esophageal Neoplasms/genetics , Telomere/metabolism , Adenocarcinoma/genetics , Aged , Anaphase/genetics , Barrett Esophagus/complications , Chromosomes, Human/genetics , Esophagus/metabolism , Flow Cytometry , Gastroesophageal Reflux/genetics , Gastroesophageal Reflux/pathology , Humans , In Situ Hybridization, Fluorescence/methods , Metaplasia/genetics , Middle Aged , Telomere/genetics
16.
Cancer Epidemiol Biomarkers Prev ; 15(3): 509-16, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16537709

ABSTRACT

BACKGROUND AND AIMS: 17p (TP53) loss of heterozygosity (LOH) has been reported to be predictive of progression from Barrett's esophagus to esophageal adenocarcinoma, but the mechanism by which TP53 LOH develops is unknown. It could be (a) DNA deletion, (b) LOH without copy number change, or (c) tetraploidy followed by genetic loss. If an alternative biomarker assay, such as fluorescence in situ hybridization (FISH), provided equivalent results, then translation to the clinic might be accelerated, because LOH genotyping is presently limited to research centers. METHODS: We evaluated mechanisms of TP53 LOH to determine if FISH and TP53 LOH provided equivalent results on the same flow-sorted samples (n = 43) representing established stages of clonal progression (diploid, diploid with TP53 LOH, aneuploid) in 19 esophagectomy specimens. RESULTS: LOH developed by all three mechanisms: 32% had DNA deletions, 32% had no copy number change, and 37% had FISH patterns consistent with a tetraploid intermediate followed by genetic loss. Thus, FISH and LOH are not equivalent (P < 0.000001). CONCLUSIONS: LOH develops by multiple chromosome mechanisms in Barrett's esophagus, all of which can be detected by genotyping. FISH cannot detect LOH without copy number change, and dual-probe FISH is required to detect the complex genetic changes associated with a tetraploid intermediate. Alternative biomarker assay development should be guided by appreciation and evaluation of the biological mechanisms generating the biomarker abnormality to detect potential sources of discordance. FISH will require validation in adequately powered longitudinal studies before implementation as a clinical diagnostic for esophageal adenocarcinoma risk prediction.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Esophageal Neoplasms/genetics , Genes, p53 , Loss of Heterozygosity , Precancerous Conditions/pathology , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Barrett Esophagus/mortality , Barrett Esophagus/pathology , Biomarkers, Tumor/analysis , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophagectomy , Esophagoscopy , Female , Humans , In Situ Hybridization, Fluorescence , Male , Neoplasm Staging , Prognosis , Retrospective Studies , Risk Assessment , Sensitivity and Specificity , Survival Rate
17.
Am J Surg Pathol ; 30(4): 423-35, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16625087

ABSTRACT

Little is known regarding the significance of esophageal biopsies that show dysplasia-like atypia limited to the bases of the crypts, without involvement of the surface epithelium in Barrett's esophagus (BE). The aim of this study was to evaluate the clinical, pathologic, immunohistochemical, and molecular characteristics of basal crypt dysplasia-like atypia (BCDA) with surface maturation in surveillance endoscopic mucosal biopsies to gain insight into its biologic significance. The Seattle Barrett's Esophagus Project is a prospective cohort study in which patients and their biopsies have been evaluated prospectively for clinical, pathologic, and molecular markers. As part of continued surveillance of the cohort, 206 consecutive BE patients were evaluated prospectively for BCDA between July 1, 2001 and August 13, 2003; 15 patients had BCDA (prevalence rate = 7.3%). These 15 patients were evaluated for clinical, pathologic, and immunohistochemical (p53 and MIB-1) features during the study period (2001-2003) as well as associations with clinical, pathologic, and molecular markers [17p(TP53) loss of heterozygosity (LOH), 9p(p16) LOH, tetraploidy, and aneuploidy] that were detected previously in the same patients in the cohort study (1983-2001). All BE patients with BCDA (male-to female ratio, 12:3; mean age, 72 years; mean length of BE, 7.0 cm; mean duration of BE, 95.1 months), except 2 (87%), had dysplasia or adenocarcinoma detected in biopsies either prior to or concurrent to the one that contained BCDA. In contrast, only 112 of 191 (59%) controls had neoplasia during the same time period (59%, P = 0.05). The difference between BCDA and controls was particularly significant with regard to the association with high-grade dysplasia (P = 0.004). Compared with adjacent nonatypical and nondysplastic (metaplastic) BE, areas of BCDA showed a significantly elevated prevalence rate of p53 positivity (60% vs. 13%, P<0.02) and a significantly elevated total crypt and basal crypt MIB-1 proliferation rate (P<0.001). Indeed, the MIB-1 proliferation rate in the basal portion of the crypts in BCDA was similar to that detected in conventional low- or high-grade dysplasia. Patients with BCDA showed a significantly increased rate of 17p(TP53) LOH (P = 0.016), aneuploidy (P = 0.004), and a trend in increased 9p(p16) LOH (P = 0.08), compared with control patients without BCDA. The clinical, pathologic, immunohistochemical, and molecular abnormalities were similar in BCDA cases that were considered low-grade versus those considered high-grade by histologic evaluation, except that high-grade cases tended to be older (79 years vs. 68 years, P = 0.06). BCDA with surface maturation, in mucosal biopsies from patients with BE, is an uncommon but significant pathologic change that shows a variety of proliferative and molecular abnormalities and has a high association with conventional dysplasia and/or adenocarcinoma. Based on these findings, BCDA warrants further investigation as a possible subtype of true dysplasia despite the morphologic appearance of surface maturation.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Esophageal Neoplasms/genetics , Gastric Mucosa/pathology , Precancerous Conditions/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Biomarkers, Tumor/metabolism , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 9/genetics , Cohort Studies , DNA, Neoplasm/analysis , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Gastric Mucosa/metabolism , Humans , Loss of Heterozygosity , Male , Middle Aged , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Prevalence , Prospective Studies , Risk Factors
18.
Hum Pathol ; 37(10): 1304-15, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16949933

ABSTRACT

We have previously demonstrated a specific pattern of mucin (MUC) core polypeptide expression in Barrett's esophagus (BE) characterized by MUC1 and MUC6 positivity in goblet cells in a proportion of cases. The aim of this study was to determine the pattern of MUC expression associated with the development and progression of dysplasia in BE. Endoscopic mucosal biopsies from 35 patients with BE (10 with no dysplasia, 6 with indefinite for dysplasia, 12 with low-grade dysplasia [LGD], and 7 biopsies with high-grade dysplasia [HGD]) were immunostained (ABC method) with antibodies against MUC1, MUC2, MUC3, MUC5AC, and MUC6. The extent and pattern of staining for each marker was evaluated in intestinalized Barrett's epithelium and in the various grades of dysplasia. For cases with dysplasia, staining was evaluated separately in nondysplastic epithelium adjacent to (<1 cm) and distant from (>2 cm) areas of dysplasia. In nondysplastic BE, MUC1, MUC2, MUC3, MUC5AC, and MUC6 stained 40%, 100%, 100%, 100%, and 90% of cases, respectively, in goblet or nongoblet columnar epithelium. With the progression of dysplasia (from metaplasia to indefinite, LGD and HGD), there was a significant decrease in expression of MUC1, MUC2, and MUC3, and alterations in the staining patterns of MUC5 and MUC6. In fact, MUC1 and MUC3 were entirely absent from all cases of HGD. Interestingly, BE-associated adenocarcinomas showed an MUC phenotype distinct from that of HGD, with expression of MUC1 and MUC3 in 47% and 67% of cases, and expression of MUC1 in a membranous pattern. There was no significant difference in MUC staining in nondysplastic BE between patients with and without dysplasia. Alterations in MUC expression occur in the progression of dysplasia in BE. However, none of these markers helps identify a subgroup of patients at increased risk for neoplasia.


Subject(s)
Adenocarcinoma/metabolism , Barrett Esophagus/metabolism , Esophageal Neoplasms/metabolism , Mucins/metabolism , Precancerous Conditions/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Barrett Esophagus/pathology , Biomarkers, Tumor/metabolism , Disease Progression , Esophageal Neoplasms/pathology , Female , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Proteins/metabolism , Peptides/metabolism , Precancerous Conditions/pathology
19.
Cancer Res ; 64(20): 7629-33, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15492292

ABSTRACT

There is debate in the literature over the relative importance of genetic instability and clonal expansion during progression to cancer. Barrett's esophagus is a uniquely suited model to investigate neoplastic progression prospectively because periodic endoscopic biopsy surveillance is recommended for early detection of esophageal adenocarcinoma. We hypothesized that expansion of clones with genetic instability would predict progression to esophageal adenocarcinoma. We measured p16 (CDKN2A/INK4A) lesions (loss of heterozygosity, mutations, and CpG island methylation), p53 (TP53) lesions (loss of heterozygosity, mutation) and ploidy abnormalities (aneuploidy, tetraploidy) within each Barrett's esophagus segment of a cohort of 267 research participants, who were followed prospectively with cancer as an outcome. We defined the size of a lesion as the fraction of cells with the lesion multiplied by the length of the Barrett's esophagus segment. A Cox proportional hazards regression indicates that the sizes of clones with p53 loss of heterozygosity (relative risk = 1.27(x) for an x cm clone; 95% confidence interval, 1.07-1.50) or ploidy abnormalities (relative risk = 1.31(x) for an x cm clone; 95% confidence interval, 1.07-1.60) predict progression to esophageal adenocarcinoma better than the mere presence of such clones (likelihood ratio test, P < 0.01). Controlling for length of the Barrett's esophagus segment had little effect. The size of a clone with a p16 lesion is not a significant predictor of esophageal adenocarcinoma when we controlled for p53 loss of heterozygosity status. The combination of clonal expansion and genetic instability is a better predictor of cancer outcome than either alone. This implies that interventions that limit expansion of genetically unstable clones may reduce risk of progression to cancer.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Aneuploidy , Cohort Studies , Disease Progression , Female , Genes, p16 , Genes, p53/genetics , Genetic Predisposition to Disease , Humans , Loss of Heterozygosity , Male , Middle Aged
20.
Cancer Epidemiol Biomarkers Prev ; 14(10): 2377-83, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16214920

ABSTRACT

Risk factors for esophageal adenocarcinoma include obesity, high fat intake, and low consumption of fruits and vegetables. This trial tested whether an intervention to reduce these risk factors in patients with Barrett esophagus, a preneoplastic condition for esophageal adenocarcinoma, could reduce biomarkers of cellular proliferation and, by inference, the risk of neoplastic progression. Eighty-seven men and women with Barrett esophagus were randomized to an intensive dietary intervention or control group. At baseline, 18 and 36 months after intervention, biopsies were obtained at 2-cm intervals throughout the length of the Barrett segment. Ki67/DNA content flow cytometry was used to assess (a) % Ki67-positive proliferating diploid G(1) cells, (b) % total Ki67-positive proliferating cells, (c) presence of aneuploidy, and (d) presence of >6% of cells in the 4N (G(2)/tetraploid) fraction of the cell cycle. We also assessed re-epithelialization and length of the Barrett segment, reflux symptoms, and medication use. The intervention effects for energy, fat, fruits and vegetables, and weight were, respectively, -314 kcal, -12.2% energy, 1.8 servings/d, and -4.0 kg at 18 months (all P < 0.005) and were smaller but remained significant at 36 months. There were no significant effects of the intervention on any biomarker of cellular proliferation. The intervention effects +/- SE for mean %G(1) Ki67+ cells were 0.98 +/- 1.58 at 18 months and 1.79 +/- 1.31 at 36 months; the relative risks (95% confidence interval) for developing >6% of cells in 4N were 0.5 (0.1-2.6) at 18 months and 0.75 (0.2-3.1) at 36 months. A single control participant developed aneuploidy. There were no significant effects on re-epithelialization, segment length, or reflux medication use. We conclude that substantial dietary change has no short-term effects on biomarkers of cellular proliferation in Barrett esophagus or on clinical observations of the Barrrett segment.


Subject(s)
Adenocarcinoma/prevention & control , Barrett Esophagus/diet therapy , Dietary Fats/administration & dosage , Esophageal Neoplasms/prevention & control , Fruit , Ki-67 Antigen/isolation & purification , Vegetables , Adenocarcinoma/etiology , Barrett Esophagus/complications , Barrett Esophagus/metabolism , Biomarkers , Esophageal Neoplasms/etiology , Female , Humans , Male , Middle Aged , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL