Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Matthew F. Buas is active.

Publication


Featured researches published by Matthew F. Buas.


Seminars in Radiation Oncology | 2013

Epidemiology and Risk Factors for Gastroesophageal Junction Tumors: Understanding the Rising Incidence of This Disease

Matthew F. Buas; Thomas L. Vaughan

Gastroesophageal (GE) junction carcinoma is a rare but often lethal condition with increasing importance as a public health problem in recent decades. Whereas diagnosis of this disease has been complicated historically by the lack of uniform classification standards, available data from the Surveillance, Epidemiology, and End Results cancer registry program in the United States show an approximate 2.5-fold increase in the incidence of GE junction adenocarcinoma from 1973 to 1992, with rates stabilizing in the past 2 decades. Similar proportional trends are observed among subgroups defined by race and gender, but rates are significantly higher in males relative to females, and in white males relative to black males. Smoking, obesity, and GE reflux disease are significant risk factors for GE junction adenocarcinoma, and may account for a substantial fraction of total disease burden. Infection with Helicobacter pylori has been associated with reduced incidence, and high dietary fiber intake has also been linked to lower disease risk. Ongoing studies continue to explore a potential role for nonsteroidal anti-inflammatory drugs in chemoprevention.


Lancet Oncology | 2016

Genome-wide association studies in oesophageal adenocarcinoma and Barrett's oesophagus: a large-scale meta-analysis

Puya Gharahkhani; Rebecca C. Fitzgerald; Thomas L. Vaughan; Claire Palles; Ines Gockel; Ian Tomlinson; Matthew F. Buas; Andrea May; Christian Gerges; Mario Anders; Jessica Becker; Nicole Kreuser; Tania Noder; Marino Venerito; Lothar Veits; Thomas Schmidt; Hendrik Manner; Claudia Schmidt; Timo Hess; Anne C. Böhmer; Jakob R. Izbicki; Arnulf H. Hölscher; Hauke Lang; Dietmar Lorenz; Brigitte Schumacher; Andreas Hackelsberger; Rupert Mayershofer; Oliver Pech; Yogesh K. Vashist; Katja Ott

Summary Background Oesophageal adenocarcinoma represents one of the fastest rising cancers in high-income countries. Barretts oesophagus is the premalignant precursor of oesophageal adenocarcinoma. However, only a few patients with Barretts oesophagus develop adenocarcinoma, which complicates clinical management in the absence of valid predictors. Within an international consortium investigating the genetics of Barretts oesophagus and oesophageal adenocarcinoma, we aimed to identify novel genetic risk variants for the development of Barretts oesophagus and oesophageal adenocarcinoma. Methods We did a meta-analysis of all genome-wide association studies of Barretts oesophagus and oesophageal adenocarcinoma available in PubMed up to Feb 29, 2016; all patients were of European ancestry and disease was confirmed histopathologically. All participants were from four separate studies within Europe, North America, and Australia and were genotyped on high-density single nucleotide polymorphism (SNP) arrays. Meta-analysis was done with a fixed-effects inverse variance-weighting approach and with a standard genome-wide significance threshold (p<5 × 10−8). We also did an association analysis after reweighting of loci with an approach that investigates annotation enrichment among genome-wide significant loci. Furthermore, the entire dataset was analysed with bioinformatics approaches—including functional annotation databases and gene-based and pathway-based methods—to identify pathophysiologically relevant cellular mechanisms. Findings Our sample comprised 6167 patients with Barretts oesophagus and 4112 individuals with oesophageal adenocarcinoma, in addition to 17 159 representative controls from four genome-wide association studies in Europe, North America, and Australia. We identified eight new risk loci associated with either Barretts oesophagus or oesophageal adenocarcinoma, within or near the genes CFTR (rs17451754; p=4·8 × 10−10), MSRA (rs17749155; p=5·2 × 10−10), LINC00208 and BLK (rs10108511; p=2·1 × 10−9), KHDRBS2 (rs62423175; p=3·0 × 10−9), TPPP and CEP72 (rs9918259; p=3·2 × 10−9), TMOD1 (rs7852462; p=1·5 × 10−8), SATB2 (rs139606545; p=2·0 × 10−8), and HTR3C and ABCC5 (rs9823696; p=1·6 × 10−8). The locus identified near HTR3C and ABCC5 (rs9823696) was associated specifically with oesophageal adenocarcinoma (p=1·6 × 10−8) and was independent of Barretts oesophagus development (p=0·45). A ninth novel risk locus was identified within the gene LPA (rs12207195; posterior probability 0·925) after reweighting with significantly enriched annotations. The strongest disease pathways identified (p<10−6) belonged to muscle cell differentiation and to mesenchyme development and differentiation. Interpretation Our meta-analysis of genome-wide association studies doubled the number of known risk loci for Barretts oesophagus and oesophageal adenocarcinoma and revealed new insights into causes of these diseases. Furthermore, the specific association between oesophageal adenocarcinoma and the locus near HTR3C and ABCC5 might constitute a novel genetic marker for prediction of the transition from Barretts oesophagus to oesophageal adenocarcinoma. Fine-mapping and functional studies of new risk loci could lead to identification of key molecules in the development of Barretts oesophagus and oesophageal adenocarcinoma, which might encourage development of advanced prevention and intervention strategies. Funding US National Cancer Institute, US National Institutes of Health, National Health and Medical Research Council of Australia, Swedish Cancer Society, Medical Research Council UK, Cambridge NIHR Biomedical Research Centre, Cambridge Experimental Cancer Medicine Centre, Else Kröner Fresenius Stiftung, Wellcome Trust, Cancer Research UK, AstraZeneca UK, University Hospitals of Leicester, University of Oxford, Australian Research Council.


Gynecologic Oncology | 2016

Identification of novel candidate plasma metabolite biomarkers for distinguishing serous ovarian carcinoma and benign serous ovarian tumors

Matthew F. Buas; Haiwei Gu; Danijel Djukovic; Jiangjiang Zhu; Charles W. Drescher; Nicole Urban; Daniel Raftery; Christopher I. Li

OBJECTIVE Serous ovarian carcinoma (OC) represents a leading cause of cancer-related death among U.S. women. Non-invasive tools have recently emerged for discriminating benign from malignant ovarian masses, but evaluation remains ongoing, without widespread implementation. In the last decade, metabolomics has matured into a new avenue for cancer biomarker development. Here, we sought to identify novel plasma metabolite biomarkers to distinguish serous ovarian carcinoma and benign serous ovarian tumor. METHODS Using liquid chromatography-mass spectrometry, we conducted global and targeted metabolite profiling of plasma isolated at the time of surgery from 50 serous OC cases and 50 serous benign controls. RESULTS Global lipidomics analysis identified 34 metabolites (of 372 assessed) differing significantly (P<0.05) between cases and controls in both training and testing sets, with 17 candidates satisfying FDR q<0.05, and two reaching Bonferroni significance. Targeted profiling of ~150 aqueous metabolites identified a single amino acid, alanine, as differentially abundant (P<0.05). A multivariate classification model built using the top four lipid metabolites achieved an estimated AUC of 0.85 (SD=0.07) based on Monte Carlo cross validation. Evaluation of a hybrid model incorporating both CA125 and lipid metabolites was suggestive of increased classification accuracy (AUC=0.91, SD=0.05) relative to CA125 alone (AUC=0.87, SD=0.07), particularly at high fixed levels of sensitivity, without reaching significance. CONCLUSIONS Our results provide insight into metabolic changes potentially correlated with the presence of serous OC versus benign ovarian tumor and suggest that plasma metabolites may help differentiate these two conditions.


PLOS ONE | 2015

MiRNA-Related SNPs and Risk of Esophageal Adenocarcinoma and Barrett’s Esophagus: Post Genome-Wide Association Analysis in the BEACON Consortium

Matthew F. Buas; Lynn Onstad; David M. Levine; Harvey A. Risch; Wong-Ho Chow; Geoffrey Liu; Rebecca C. Fitzgerald; Leslie Bernstein; Weimin Ye; Nigel C. Bird; Yvonne Romero; Alan G. Casson; Douglas A. Corley; Nicholas J. Shaheen; Anna H. Wu; Marilie D. Gammon; Brian J. Reid; Laura J. Hardie; Ulrike Peters; David C. Whiteman; Thomas L. Vaughan

Incidence of esophageal adenocarcinoma (EA) has increased substantially in recent decades. Multiple risk factors have been identified for EA and its precursor, Barrett’s esophagus (BE), such as reflux, European ancestry, male sex, obesity, and tobacco smoking, and several germline genetic variants were recently associated with disease risk. Using data from the Barrett’s and Esophageal Adenocarcinoma Consortium (BEACON) genome-wide association study (GWAS) of 2,515 EA cases, 3,295 BE cases, and 3,207 controls, we examined single nucleotide polymorphisms (SNPs) that potentially affect the biogenesis or biological activity of microRNAs (miRNAs), small non-coding RNAs implicated in post-transcriptional gene regulation, and deregulated in many cancers, including EA. Polymorphisms in three classes of genes were examined for association with risk of EA or BE: miRNA biogenesis genes (157 SNPs, 21 genes); miRNA gene loci (234 SNPs, 210 genes); and miRNA-targeted mRNAs (177 SNPs, 158 genes). Nominal associations (P<0.05) of 29 SNPs with EA risk, and 25 SNPs with BE risk, were observed. None remained significant after correction for multiple comparisons (FDR q>0.50), and we did not find evidence for interactions between variants analyzed and two risk factors for EA/BE (smoking and obesity). This analysis provides the most extensive assessment to date of miRNA-related SNPs in relation to risk of EA and BE. While common genetic variants within components of the miRNA biogenesis core pathway appear unlikely to modulate susceptibility to EA or BE, further studies may be warranted to examine potential associations between unassessed variants in miRNA genes and targets with disease risk.


Cancer Epidemiology, Biomarkers & Prevention | 2015

A Newly Identified Susceptibility Locus near FOXP1 Modifies the Association of Gastroesophageal Reflux with Barrett's Esophagus

James Y. Dai; Jean de Dieu Tapsoba; Matthew F. Buas; Lynn Onstad; David M. Levine; Harvey A. Risch; Wong Ho Chow; Leslie Bernstein; Weimin Ye; Jesper Lagergren; Nigel C. Bird; Douglas A. Corley; Nicholas J. Shaheen; Anna H. Wu; Brian J. Reid; Laura J. Hardie; David C. Whiteman; Thomas L. Vaughan

Background: Important risk factors for esophageal adenocarcinoma and its precursor, Barretts esophagus, include gastroesophageal reflux disease, obesity, and cigarette smoking. Recently, genome-wide association studies have identified seven germline single-nucleotide polymorphisms (SNP) that are associated with risk of Barretts esophagus and esophageal adenocarcinoma. Whether these genetic susceptibility loci modify previously identified exposure–disease associations is unclear. Methods: We analyzed exposure and genotype data from the BEACON Consortium discovery phase GWAS, which included 1,516 esophageal adenocarcinoma case patients, 2,416 Barretts esophagus case patients, and 2,187 control participants. We examined the seven newly identified susceptibility SNPs for interactions with body mass index, smoking status, and report of weekly heartburn or reflux. Logistic regression models were used to estimate ORs for these risk factors stratified by SNP genotype, separately for Barretts esophagus and esophageal adenocarcinoma. Results: The odds ratio for Barretts esophagus associated with at least weekly heartburn or reflux varied significantly with the presence of at least one minor allele of rs2687201 (nominal P = 0.0005, FDR = 0.042). ORs (95% CIs) for weekly heartburn or reflux among participants with 0, 1, or 2 minor alleles of rs2687201 were 6.17 (4.91–7.56), 3.56 (2.85–4.44), and 3.97 (2.47–6.37), respectively. No statistically significant interactions were observed for smoking status and body mass index. Conclusion: Reflux symptoms are more strongly associated with Barretts esophagus risk among persons homozygous for the major allele of rs2687201, which lies approximately 75 kb downstream of the transcription factor gene FOXP1. Impact: The novel gene–exposure interaction discovered in this study provides new insights into the etiology of esophageal adenocarcinoma. Cancer Epidemiol Biomarkers Prev; 24(11); 1739–47. ©2015 AACR.


PLOS ONE | 2015

SNP regulation of microRNA expression and subsequent colon cancer risk

Lila E. Mullany; Roger K. Wolff; Jennifer S. Herrick; Matthew F. Buas; Martha L. Slattery

Introduction MicroRNAs (miRNAs) regulate messenger RNAs (mRNAs) and as such have been implicated in a variety of diseases, including cancer. MiRNAs regulate mRNAs through binding of the miRNA 5’ seed sequence (~7–8 nucleotides) to the mRNA 3’ UTRs; polymorphisms in these regions have the potential to alter miRNA-mRNA target associations. SNPs in miRNA genes as well as miRNA-target genes have been proposed to influence cancer risk through altered miRNA expression levels. Methods MiRNA-SNPs and miRNA-target gene-SNPs were identified through the literature. We used SNPs from Genome-Wide Association Study (GWAS) data that were matched to individuals with miRNA expression data generated from an Agilent platform for colon tumor and non-tumor paired tissues. These samples were used to evaluate 327 miRNA-SNP pairs for associations between SNPs and miRNA expression levels as well as for SNP associations with colon cancer. Results Twenty-two miRNAs expressed in non-tumor tissue were significantly different by genotype and 21 SNPs were associated with altered tumor/non-tumor differential miRNA expression across genotypes. Two miRNAs were associated with SNP genotype for both non-tumor and tumor/non-tumor differential expression. Of the 41 miRNAs significantly associated with SNPs all but seven were significantly differentially expressed in colon tumor tissue. Two of the 41 SNPs significantly associated with miRNA expression levels were associated with colon cancer risk: rs8176318 (BRCA1), ORAA 1.31 95% CI 1.01, 1.78, and rs8905 (PRKAR1A), ORGG 2.31 95% CI 1.11, 4.77. Conclusion Of the 327 SNPs identified in the literature as being important because of their potential regulation of miRNA expression levels, 12.5% had statistically significantly associations with miRNA expression. However, only two of these SNPs were significantly associated with colon cancer.


Gut | 2017

Germline variation in inflammation-related pathways and risk of Barrett's oesophagus and oesophageal adenocarcinoma

Matthew F. Buas; Qianchuan He; Lisa G. Johnson; Lynn Onstad; David M. Levine; Aaron P. Thrift; Puya Gharahkhani; Claire Palles; Jesper Lagergren; Rebecca C. Fitzgerald; Weimin Ye; Carlos Caldas; Nigel C. Bird; Nicholas J. Shaheen; Leslie Bernstein; Marilie D. Gammon; Anna H. Wu; Laura J. Hardie; Paul Pharoah; Geoffrey Liu; Prassad Iyer; Douglas A. Corley; Harvey A. Risch; Wong-Ho Chow; Hans Prenen; Laura Chegwidden; Sharon Love; Stephen Attwood; Paul Moayyedi; David MacDonald

Objective Oesophageal adenocarcinoma (OA) incidence has risen sharply in Western countries over recent decades. Local and systemic inflammation is considered an important contributor to OA pathogenesis. Established risk factors for OA and its precursor, Barretts oesophagus (BE), include symptomatic reflux, obesity and smoking. The role of inherited genetic susceptibility remains an area of active investigation. Here, we explore whether germline variation related to inflammatory processes influences susceptibility to BE/OA. Design We used data from a genomewide association study of 2515 OA cases, 3295 BE cases and 3207 controls. Our analysis included 7863 single-nucleotide polymorphisms (SNPs) in 449 genes assigned to five pathways: cyclooxygenase (COX), cytokine signalling, oxidative stress, human leucocyte antigen and nuclear factor-κB. A principal components-based analytic framework was employed to evaluate pathway-level and gene-level associations with disease risk. Results We identified a significant signal for the COX pathway in relation to BE risk (p=0.0059, false discovery rate q=0.03), and in gene-level analyses found an association with microsomal glutathione-S-transferase 1 (MGST1); (p=0.0005, q=0.005). Assessment of 36 MGST1 SNPs identified 14 variants associated with elevated BE risk (q<0.05). Four of these were subsequently confirmed (p<5.5×10−5) in a meta-analysis encompassing an independent set of 1851 BE cases and 3496 controls, and are known strong expression quantitative trait loci for MGST1. Three such variants were associated with similar elevations in OA risk. Conclusions This study provides the most comprehensive evaluation of inflammation-related germline variation in relation to risk of BE/OA and suggests that variants in MGST1 influence disease susceptibility.


Metabolomics | 2017

Candidate serum metabolite biomarkers for differentiating gastroesophageal reflux disease, Barrett’s esophagus, and high-grade dysplasia/esophageal adenocarcinoma

Matthew F. Buas; Haiwei Gu; Danijel Djukovic; Jiangjiang Zhu; Lynn Onstad; Brian J. Reid; Daniel Raftery; Thomas L. Vaughan

Introduction/objectivesIncidence of esophageal adenocarcinoma (EA), an often fatal cancer, has increased sharply over recent decades. Several important risk factors (reflux, obesity, smoking) have been identified for EA and its precursor, Barrett’s esophagus (BE), but a key challenge remains in identifying individuals at highest risk, since most with reflux do not develop BE, and most with BE do not progress to cancer. Metabolomics represents an emerging approach for identifying novel biomarkers associated with cancer development.MethodsWe used targeted liquid chromatography-mass spectrometry (LC-MS) to profile 57 metabolites in 322 serum specimens derived from individuals with gastroesophageal reflux disease (GERD), BE, high-grade dysplasia (HGD), or EA, drawn from two well-annotated epidemiologic parent studies.ResultsMultiple metabolites differed significantly (P < 0.05) between BE versus GERD (n = 9), and between HGD/EA versus BE (n = 4). Several top candidates (FDR q ≤ 0.15), including urate, homocysteine, and 3-nitrotyrosine, are linked to inflammatory processes, which may contribute to BE/EA pathogenesis. Multivariate modeling achieved moderate discrimination between HGD/EA and BE (AUC = 0.75), with less pronounced separation for BE versus GERD (AUC = 0.64).ConclusionSerum metabolite differences can be detected between individuals with GERD versus BE, and between those with BE versus HGD/EA, and may help differentiate patients at different stages of progression to EA.


American Journal of Human Genetics | 2016

Constrained Score Statistics Identify Genetic Variants Interacting with Multiple Risk Factors in Barrett’s Esophagus

James Y. Dai; Jean de Dieu Tapsoba; Matthew F. Buas; Wong Ho Chow; Nicholas J. Shaheen; Lesley Anderson; Douglas A. Corley; Marilie D. Gammon; Laura J. Hardie; Jesper Lagergren; David C. Whiteman; Harvey A. Risch; Thomas L. Vaughan

Few gene-environment interactions (G × E) have been discovered in cancer epidemiology thus far, in part due to the large number of possible G × E to be investigated and inherent low statistical power of traditional analytic methods for discovering G × E. We consider simultaneously testing for interactions between several related exposures and a genetic variant in a genome-wide study. To improve power, constrained testing strategies are proposed for multivariate gene-environment interactions at two levels: interactions that have the same direction (one-sided or bidirectional hypotheses) or are proportional to respective exposure main effects (a variant of Tukeys one-degree test). Score statistics were developed to expedite the genome-wide computation. We conducted extensive simulations to evaluate validity and power performance of the proposed statistics, applied them to the genetic and environmental exposure data for esophageal adenocarcinoma and Barretts esophagus from the Barretts Esophagus and Esophageal Adenocarcinoma Consortium (BEACON), and discovered three loci simultaneously interacting with gastresophageal reflux, obesity, and tobacco smoking with genome-wide significance. These findings deepen understanding of the genetic and environmental architecture of Barretts esophagus and esophageal adenocarcinoma.


Cancer Research | 2013

Abstract 2561: MiRNA-related SNPs and risk of esophageal adenocarcinoma and Barrett's esophagus: post genome-wide association analysis in the BEACON consortium.

Matthew F. Buas; Lynn Onstad; David K. Levine; Thomas L. Vaughan; David C. Whiteman

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC The incidence of esophageal adenocarcinoma (EA) has increased significantly in recent decades. Although several major risk factors have been identified for EA and its precursor, Barretts esophagus (BE), including reflux, Caucasian race, male gender, obesity, and smoking, only limited knowledge exists regarding the role of inherited genetic variation and its interplay with environmental factors. The Barretts and Esophageal Adenocarcinoma Consortium (BEACON) recently completed a genome-wide association study (GWAS) of 1,517 EA cases, 2,416 BE cases, and 2,187 controls. Using this dataset, we examined single nucleotide polymorphisms (SNPs) that potentially affect the biogenesis or biological activity of microRNAs (miRNAs), a class of small non-coding RNAs that regulate post-transcriptional gene expression and are deregulated in many cancers, including EA. Polymorphisms in three classes of genes were evaluated for their association with risk of EA or BE: 1) miRNA biogenesis genes (157 SNPs), 2) miRNA gene loci (234 SNPs), and 3) miRNA-targeted mRNAs (179 SNPs). 28 SNPs were significantly associated (P 30% reduced risk of EA (OR 0.68, 95% CI 0.55-0.83), which remained significant after correction for multiple comparisons (adjusted P=0.04). This inverse association became more pronounced with increasing pack-years (P-interaction=0.02). Alterations in disease risk associated with other polymorphisms ranged from 10-40%. This study provides the most extensive assessment to date of miRNA-related SNPs in relation to risk of EA and BE and suggests that genetic variation in this pathway may influence disease susceptibility and interact with known risk factors. Citation Format: Matthew F. Buas, Lynn Onstad, David Levine, Thomas L. Vaughan, David C. Whiteman, BEACON Consortium Investigators. MiRNA-related SNPs and risk of esophageal adenocarcinoma and Barretts esophagus: post genome-wide association analysis in the BEACON consortium. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2561. doi:10.1158/1538-7445.AM2013-2561

Collaboration


Dive into the Matthew F. Buas's collaboration.

Top Co-Authors

Avatar

Thomas L. Vaughan

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Lynn Onstad

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Brian J. Reid

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicholas J. Shaheen

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David C. Whiteman

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Anna H. Wu

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge