Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
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
2.
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
3.
Gastroenterology ; 159(6): 2065-2076.e1, 2020 12.
Article in English | MEDLINE | ID: mdl-32918910

ABSTRACT

BACKGROUND & AIMS: Esophageal adenocarcinoma (EA) and its premalignant lesion, Barrett's esophagus (BE), are characterized by a strong and yet unexplained male predominance (with a male-to-female ratio in EA incidence of up to 6:1). Genome-wide association studies (GWAS) have identified more than 20 susceptibility loci for these conditions. However, potential sex differences in genetic associations with BE/EA remain largely unexplored. METHODS: Given strong genetic overlap, BE and EA cases were combined into a single case group for analysis. These were compared with population-based controls. We performed sex-specific GWAS of BE/EA in 3 separate studies and then used fixed-effects meta-analysis to provide summary estimates for >9 million variants for male and female individuals. A series of downstream analyses were conducted separately in male and female individuals to identify genes associated with BE/EA and the genetic correlations between BE/EA and other traits. RESULTS: We included 6758 male BE/EA cases, 7489 male controls, 1670 female BE/EA cases, and 6174 female controls. After Bonferroni correction, our meta-analysis of sex-specific GWAS identified 1 variant at chromosome 6q11.1 (rs112894788, KHDRBS2-MTRNR2L9, PBONF = .039) that was statistically significantly associated with BE/EA risk in male individuals only, and 1 variant at chromosome 8p23.1 (rs13259457, PRSS55-RP1L1, PBONF = 0.057) associated, at borderline significance, with BE/EA risk in female individuals only. We also observed strong genetic correlations of BE/EA with gastroesophageal reflux disease in male individuals and obesity in female individuals. CONCLUSIONS: The identified novel sex-specific variants associated with BE/EA could improve the understanding of the genetic architecture of the disease and the reasons for the male predominance.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Biomarkers, Tumor/genetics , Esophageal Neoplasms/genetics , Genetic Predisposition to Disease , Adenocarcinoma/epidemiology , Barrett Esophagus/epidemiology , Case-Control Studies , Esophageal Neoplasms/epidemiology , Eye Proteins/genetics , Female , Gastroesophageal Reflux/epidemiology , Gastroesophageal Reflux/genetics , Genetic Loci , Genome-Wide Association Study , Humans , Male , Obesity/epidemiology , Obesity/genetics , Polymorphism, Single Nucleotide , RNA-Binding Proteins/genetics , Risk Assessment , Risk Factors , Serine Endopeptidases/genetics , Sex Factors
4.
Clin Gastroenterol Hepatol ; 18(12): 2701-2709.e3, 2020 11.
Article in English | MEDLINE | ID: mdl-31756444

ABSTRACT

BACKGROUND & AIMS: Esophageal adenocarcinoma (EAC) occurs most frequently in men. We performed a Mendelian randomization analysis to investigate whether genetic factors that regulate levels of sex hormones are associated with risk of EAC or Barrett's esophagus (BE). METHODS: We conducted a Mendelian randomization analysis using data from patients with EAC (n = 2488) or BE (n = 3247) and control participants (n = 2127), included in international consortia of genome-wide association studies in Australia, Europe, and North America. Genetic risk scores or single-nucleotide variants were used as instrumental variables for 9 specific sex hormones. Logistic regression provided odds ratios (ORs) with 95% CIs. RESULTS: Higher genetically predicted levels of follicle-stimulating hormones were associated with increased risks of EAC and/or BE in men (OR, 1.14 per allele increase; 95% CI, 1.01-1.27) and in women (OR, 1.28; 95% CI, 1.03-1.59). Higher predicted levels of luteinizing hormone were associated with a decreased risk of EAC in men (OR, 0.92 per SD increase; 95% CI, 0.87-0.99) and in women (OR, 0.93; 95% CI, 0.79-1.09), and decreased risks of BE (OR, 0.88; 95% CI, 0.77-0.99) and EAC and/or BE (OR, 0.89; 95% CI, 0.79-1.00) in women. We found no clear associations for other hormones studied, including sex hormone-binding globulin, dehydroepiandrosterone sulfate, testosterone, dihydrotestosterone, estradiol, progesterone, or free androgen index. CONCLUSIONS: In a Mendelian randomization analysis of data from patients with EAC or BE, we found an association between genetically predicted levels of follicle-stimulating and luteinizing hormones and risk of BE and EAC.


Subject(s)
Adenocarcinoma , Barrett Esophagus , Esophageal Neoplasms , Adenocarcinoma/epidemiology , Barrett Esophagus/genetics , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/genetics , Female , Genome-Wide Association Study , Gonadal Steroid Hormones , Humans , Male , Risk Factors
5.
Gastroenterology ; 154(5): 1273-1281.e3, 2018 04.
Article in English | MEDLINE | ID: mdl-29247777

ABSTRACT

BACKGROUND & AIMS: We developed comprehensive models to determine risk of Barrett's esophagus (BE) or esophageal adenocarcinoma (EAC) based on genetic and non-genetic factors. METHODS: We used pooled data from 3288 patients with BE, 2511 patients with EAC, and 2177 individuals without either (controls) from participants in the international Barrett's and EAC consortium as well as the United Kingdom's BE gene study and stomach and esophageal cancer study. We collected data on 23 genetic variants associated with risk for BE or EAC, and constructed a polygenic risk score (PRS) for cases and controls by summing the risk allele counts for the variants weighted by their natural log-transformed effect estimates (odds ratios) extracted from genome-wide association studies. We also collected data on demographic and lifestyle factors (age, sex, smoking, body mass index, use of nonsteroidal anti-inflammatory drugs) and symptoms of gastroesophageal reflux disease (GERD). Risk models with various combinations of non-genetic factors and the PRS were compared for their accuracy in identifying patients with BE or EAC using the area under the receiver operating characteristic curve (AUC) analysis. RESULTS: Individuals in the highest quartile of risk, based on genetic factors (PRS), had a 2-fold higher risk of BE (odds ratio, 2.22; 95% confidence interval, 1.89-2.60) or EAC (odds ratio, 2.46; 95% confidence interval, 2.07-2.92) than individual in the lowest quartile of risk based on PRS. Risk models developed based on only demographic or lifestyle factors or GERD symptoms identified patients with BE or EAC with AUC values ranging from 0.637 to 0.667. Combining data on demographic or lifestyle factors with data on GERD symptoms identified patients with BE with an AUC of 0.793 and patients with EAC with an AUC of 0.745. Including PRSs with these data only minimally increased the AUC values for BE (to 0.799) and EAC (to 0.754). Including the PRSs in the model developed based on non-genetic factors resulted in a net reclassification improvement for BE of 3.0% and for EAC of 5.6%. CONCLUSIONS: We used data from 3 large databases of patients from studies of BE or EAC to develop a risk prediction model based on genetic, clinical, and demographic/lifestyle factors. We identified a PRS that increases discrimination and net reclassification of individuals with vs without BE and EAC. However, the absolute magnitude of improvement is not sufficient to justify its clinical use.


Subject(s)
Adenocarcinoma/epidemiology , Adenocarcinoma/genetics , Barrett Esophagus/epidemiology , Barrett Esophagus/genetics , Decision Support Techniques , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/genetics , Models, Genetic , Polymorphism, Single Nucleotide , Adenocarcinoma/diagnosis , Area Under Curve , Australia/epidemiology , Barrett Esophagus/diagnosis , Case-Control Studies , Databases, Factual , Esophageal Neoplasms/diagnosis , Europe/epidemiology , Female , Gene-Environment Interaction , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Life Style , Logistic Models , Male , Middle Aged , Molecular Epidemiology , Multifactorial Inheritance , North America/epidemiology , Odds Ratio , Phenotype , Predictive Value of Tests , ROC Curve , Risk Assessment , Risk Factors
6.
Clin Gastroenterol Hepatol ; 17(11): 2227-2235.e1, 2019 10.
Article in English | MEDLINE | ID: mdl-30716477

ABSTRACT

BACKGROUND & AIMS: Epidemiology studies of circulating concentrations of 25 hydroxy vitamin D (25(OH)D) and risk of esophageal adenocarcinoma (EAC) have produced conflicting results. We conducted a Mendelian randomization study to determine the associations between circulating concentrations of 25(OH)D and risks of EAC and its precursor, Barrett's esophagus (BE). METHODS: We conducted a Mendelian randomization study using a 2-sample (summary data) approach. Six single-nucleotide polymorphisms (SNPs; rs3755967, rs10741657, rs12785878, rs10745742, rs8018720, and rs17216707) associated with circulating concentrations of 25(OH)D were used as instrumental variables. We collected data from 6167 patients with BE, 4112 patients with EAC, and 17,159 individuals without BE or EAC (controls) participating in the Barrett's and Esophageal Adenocarcinoma Consortium, as well as studies from Bonn, Germany, and Cambridge and Oxford, United Kingdom. Analyses were performed separately for BE and EAC. RESULTS: Overall, we found no evidence for an association between genetically estimated 25(OH)D concentration and risk of BE or EAC. The odds ratio per 20 nmol/L increase in genetically estimated 25(OH)D concentration for BE risk estimated by combining the individual SNP association using inverse variance weighting was 1.21 (95% CI, 0.77-1.92; P = .41). The odds ratio for EAC risk, estimated by combining the individual SNP association using inverse variance weighting, was 0.68 (95% CI, 0.39-1.19; P = .18). CONCLUSIONS: In a Mendelian randomization study, we found that low genetically estimated 25(OH)D concentrations were not associated with risk of BE or EAC.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Esophageal Neoplasms/genetics , Mendelian Randomization Analysis/methods , Polymorphism, Single Nucleotide , Risk Assessment , Vitamin D/blood , Adenocarcinoma/blood , Adenocarcinoma/epidemiology , Barrett Esophagus/blood , Barrett Esophagus/epidemiology , Biomarkers, Tumor/blood , DNA, Neoplasm/genetics , Esophageal Neoplasms/blood , Esophageal Neoplasms/epidemiology , Europe/epidemiology , Female , Humans , Male , Morbidity , North America/epidemiology , Risk Factors
7.
Environ Sci Technol ; 53(1): 50-59, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30485747

ABSTRACT

The "4 per mil" initiative recognizes the pivotal role of soil in carbon resequestration. The need for evidence to substantiate the influence of agricultural practices on chemical nature of soil carbon and microbial biodiversity has become a priority. However, owing to the molecular complexity of soil dissolved organic matter (DOM), specific linkages to microbial biodiversity have eluded researchers. Here, we characterized the chemodiversity of soil DOM, assessed the variation of soil bacterial community composition (BCC), and identified specific linkages between DOM traits and BCC. Sustained organic carbon amendment significantly ( P < 0.05) increased total organic matter reservoirs, resulted in higher chemodiversity of DOM and emergence of recalcitrant moieties (H/C < 1.5). In the meantime, sustained organic carbon amendment shaped the BCC to a more eutrophic state while long-term chemical fertilization directed the BCC toward an oligotrophic state. Meanwhile, higher connectivity and complexity were observed in organic carbon amendment by DOM-BCC network analysis, indicating that soil microbes tended to have more interaction with DOM molecules after organic matter inputs. These results highlight the potential for organic carbon amendments to not only build soil carbon stocks and increase their resilience but also mediate the functional state of soil bacterial communities.


Subject(s)
Microbiota , Soil , Agriculture , Biodiversity , Carbon
8.
Clin Gastroenterol Hepatol ; 16(10): 1598-1606.e4, 2018 10.
Article in English | MEDLINE | ID: mdl-29551738

ABSTRACT

BACKGROUND & AIMS: Genome-wide association studies (GWAS) have identified more than 20 susceptibility loci for esophageal adenocarcinoma (EA) and Barrett's esophagus (BE). However, variants in these loci account for a small fraction of cases of EA and BE. Genetic factors might interact with environmental factors to affect risk of EA and BE. We aimed to identify single nucleotide polymorphisms (SNPs) that may modify the associations of body mass index (BMI), smoking, and gastroesophageal reflux disease (GERD), with risks of EA and BE. METHODS: We collected data on single BMI measurements, smoking status, and symptoms of GERD from 2284 patients with EA, 3104 patients with BE, and 2182 healthy individuals (controls) participating in the Barrett's and Esophageal Adenocarcinoma Consortium GWAS, the UK Barrett's Esophagus Gene Study, and the UK Stomach and Oesophageal Cancer Study. We analyzed 993,501 SNPs in DNA samples of all study subjects. We used standard case-control logistic regression to test for gene-environment interactions. RESULTS: For EA, rs13429103 at chromosome 2p25.1, near the RNF144A-LOC339788 gene, showed a borderline significant interaction with smoking status (P = 2.18×10-7). Ever smoking was associated with an almost 12-fold increase in risk of EA among individuals with rs13429103-AA genotype (odds ratio=11.82; 95% CI, 4.03-34.67). Three SNPs (rs12465911, rs2341926, rs13396805) at chromosome 2q23.3, near the RND3-RBM43 gene, interacted with GERD symptoms (P = 1.70×10-7, P = 1.83×10-7, and P = 3.58×10-7, respectively) to affect risk of EA. For BE, rs491603 at chromosome 1p34.3, near the EIF2C3 gene, and rs11631094 at chromosome 15q14, at the SLC12A6 gene, interacted with BMI (P = 4.44×10-7) and pack-years of smoking history (P = 2.82×10-7), respectively. CONCLUSION: The associations of BMI, smoking, and GERD symptoms with risks of EA and BE appear to vary with SNPs at chromosomes 1, 2, and 15. Validation of these suggestive interactions is warranted.


Subject(s)
Adenocarcinoma/epidemiology , Barrett Esophagus/complications , Barrett Esophagus/epidemiology , Environmental Exposure , Esophageal Neoplasms/epidemiology , Genetic Predisposition to Disease , Adenocarcinoma/etiology , Aged , Esophageal Neoplasms/etiology , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Risk Assessment , United Kingdom/epidemiology
9.
Environ Geochem Health ; 39(6): 1221-1243, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28455820

ABSTRACT

Excessive transport of fine sediment, and its associated pollutants, can cause detrimental impacts in aquatic environments. It is therefore important to perform accurate sediment source apportionment to identify hot spots of soil erosion. Various tracers have been adopted, often in combination, to identify sediment source type and its spatial origin; these include fallout radionuclides, geochemical tracers, mineral magnetic properties and bulk and compound-specific stable isotopes. In this review, the applicability of these techniques to particular settings and their advantages and limitations are reviewed. By synthesizing existing approaches, that make use of multiple tracers in combination with measured changes of channel geomorphological attributes, an integrated analysis of tracer profiles in deposited sediments in lakes and reservoirs can be made. Through a multi-scale approach for fine sediment tracking, temporal changes in soil erosion and sediment load can be reconstructed and the consequences of changing catchment practices evaluated. We recommend that long-term, as well as short-term, monitoring of riverine fine sediment and corresponding surface and subsurface sources at nested sites within a catchment are essential. Such monitoring will inform the development and validation of models for predicting dynamics of fine sediment transport as a function of hydro-climatic and geomorphological controls. We highlight that the need for monitoring is particularly important for hilly catchments with complex and changing land use. We recommend that research should be prioritized for sloping farmland-dominated catchments.


Subject(s)
Geologic Sediments/chemistry , Environmental Monitoring/methods , Fresh Water/chemistry , Models, Theoretical , Soil/chemistry , Soil Pollutants/analysis , Time Factors , Uncertainty
10.
J Environ Sci (China) ; 62: 39-48, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29289291

ABSTRACT

Understanding antibiotic biodegradation is important to the appreciation of their fate and removal from the environment. In this research an Isotope Ratio Mass Spectrometry (IRMS) method was developed to evaluate the extent of biodegradation of the antibiotic, sulphanilamide, in contaminated groundwater. Results indicted an enrichment in δ13C of 8.44‰ from -26.56 (at the contaminant source) to -18.12‰ (300m downfield of the source). These results confirm reductions in sulphanilamide concentrations (from 650 to 10mg/L) across the contaminant plume to be attributable to biodegradation (56%) vs. other natural attenuation processes, such as dilution or dispersion (42%). To understand the controls on sulphanilamide degradation ex-situ microcosms assessed the influence of sulphanilamide concentration, redox conditions and an alternative carbon source. Results indicated, high levels of anaerobic capacity (~50% mineralisation) to degrade sulphanilamide under high (263mg/L), moderate (10mg/L) and low (0.02mg/L) substrate concentrations. The addition of electron acceptors; nitrate and sulphate, did not significantly enhance the capacity of the groundwater to anaerobically biodegrade sulphanilamide. Interestingly, where alternative carbon sources were present, the addition of nitrate and sulphate inhibited sulphanilamide biodegradation. These results suggest, under in-situ conditions, when a preferential carbon source was available for biodegradation, sulphanilamide could be acting as a nitrogen and/or sulphur source. These findings are important as they highlight sulphanilamide being used as a carbon and a putative nitrogen and sulphur source, under prevailing iron reducing conditions.


Subject(s)
Groundwater/chemistry , Sulfanilamides/analysis , Water Pollutants, Chemical/analysis , Biodegradation, Environmental , Calcium Carbonate , Sulfanilamide , Water Microbiology
11.
J Cell Physiol ; 231(9): 1870-5, 2016 09.
Article in English | MEDLINE | ID: mdl-26505642

ABSTRACT

Many cancers offer an extended window of opportunity for early detection and therapeutic intervention that could lead to a reduction in cause-specific mortality. The pursuit of early detection in screening settings has resulted in decreased incidence and mortality for some cancers (e.g., colon and cervical cancers), and increased incidence with only modest or no effect on cause-specific mortality in others (e.g., breast and prostate). Whereas highly sensitive screening technologies are better at detecting a number of suspected "cancers" that are indolent and likely to remain clinically unimportant in the lifetime of a patient, defined as overdiagnosis, they often miss cancers that are aggressive and tend to present clinically between screenings, known as interval cancers. Unrecognized overdiagnosis leads to overtreatment with its attendant (often long-lasting) side effects, anxiety, and substantial financial harm. Existing methods often cannot differentiate indolent lesions from aggressive ones or understand the dynamics of neoplastic progression. To correctly identify the population that would benefit the most from screening and identify the lesions that would benefit most from treatment, the evolving genomic and molecular profiles of individual cancers during the clinical course of progression or indolence must be investigated, while taking into account an individual's genetic susceptibility, clinical and environmental risk factors, and the tumor microenvironment. Practical challenges lie not only in the lack of access to tissue specimens that are appropriate for the study of natural history, but also in the absence of targeted research strategies. This commentary summarizes the recommendations from a diverse group of scientists with expertise in basic biology, translational research, clinical research, statistics, and epidemiology and public health professionals convened to discuss research directions. J. Cell. Physiol. 231: 1870-1875, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Early Detection of Cancer , Genetic Predisposition to Disease/genetics , Mass Screening , Medical Overuse/prevention & control , Neoplasms/diagnosis , Animals , Humans , Neoplasms/genetics , Risk Factors
12.
Gastroenterology ; 149(5): 1142-1152.e3, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26208895

ABSTRACT

Beginning in the 1980s, an alarming rise in the incidence of esophageal adenocarcinoma (EA) led to screening of patients with reflux to detect Barrett's esophagus (BE) and surveillance of BE to detect early EA. This strategy, based on linear progression disease models, resulted in selective detection of BE that does not progress to EA over a lifetime (overdiagnosis) and missed BE that rapidly progresses to EA (underdiagnosis). Here we review the historical thought processes that resulted in this undesired outcome and the transformation in our understanding of genetic and evolutionary principles governing neoplastic progression that has come from application of modern genomic technologies to cancers and their precursors. This new synthesis provides improved strategies for prevention and early detection of EA by addressing the environmental and mutational processes that can determine "windows of opportunity" in time to detect rapidly progressing BE and distinguish it from slowly or nonprogressing BE.


Subject(s)
Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/prevention & control , Barrett Esophagus/pathology , Early Detection of Cancer/methods , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/genetics , Esophageal Neoplasms/prevention & control , Genomics/trends , Barrett Esophagus/etiology , Disease Progression , Early Detection of Cancer/trends , Environment , Gastroesophageal Reflux/complications , Genetic Predisposition to Disease , Genomics/methods , Humans , Mutation
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.
Nat Rev Cancer ; 6(12): 924-35, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17109012

ABSTRACT

Neoplasms are microcosms of evolution. Within a neoplasm, a mosaic of mutant cells compete for space and resources, evade predation by the immune system and can even cooperate to disperse and colonize new organs. The evolution of neoplastic cells explains both why we get cancer and why it has been so difficult to cure. The tools of evolutionary biology and ecology are providing new insights into neoplastic progression and the clinical control of cancer.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Evolution, Molecular , Gene Expression Regulation, Neoplastic , Animals , Cell Communication , Cell Proliferation , Cell Survival , Disease Progression , Drug Resistance, Neoplasm/genetics , Genetic Drift , Humans , Models, Genetic , Mutation , Neoplasm Invasiveness , Neoplasms/genetics , Neoplastic Stem Cells/pathology , Selection, Genetic
15.
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
16.
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
17.
Clin Gastroenterol Hepatol ; 12(10): 1667-76.e1, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24530603

ABSTRACT

BACKGROUND & AIMS: Risks for some cancers increase with height. We investigated the relationship between height and risk of esophageal adenocarcinoma (EAC) and its precursor, Barrett's esophagus (BE). METHODS: We analyzed epidemiologic and genome-wide genomic data from individuals of European ancestry in the Barrett's and Esophageal Adenocarcinoma Consortium, from 999 cases of EAC, 2061 cases of BE, and 2168 population controls. Multivariable logistic regression was used to estimate odds ratios (OR) and 95% confidence intervals (95% CI) for associations between height and risks of EAC and BE. We performed a Mendelian randomization analysis to estimate an unconfounded effect of height on EAC and BE using a genetic risk score derived from 243 genetic variants associated with height as an instrumental variable. RESULTS: Height was associated inversely with EAC (per 10-cm increase in height: OR, 0.70; 95% CI, 0.62-0.79 for men and OR, 0.57; 95% CI 0.40-0.80 for women) and BE (per 10-cm increase in height: OR, 0.69; 95% CI, 0.62-0.77 for men and OR, 0.61; 95% CI, 0.48-0.77 for women). The risk estimates were consistent across strata of age, education level, smoking, gastroesophageal reflux symptoms, body mass index, and weight. Mendelian randomization analysis yielded results quantitatively similar to those from the conventional epidemiologic analysis. CONCLUSIONS: Height is associated inversely with risks of EAC and BE. Results from the Mendelian randomization study showed that the inverse association observed did not result from confounding factors. Mechanistic studies of the effect of height on EAC and BE are warranted; height could have utility in clinical risk stratification.


Subject(s)
Adenocarcinoma/epidemiology , Body Height , Esophageal Neoplasms/epidemiology , Adult , Aged , Aged, 80 and over , Barrett Esophagus/epidemiology , Europe/epidemiology , Female , Humans , Male , Mendelian Randomization Analysis , Middle Aged , Risk Factors
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL