Skip to main content
Erschienen in: Digestive Diseases and Sciences 1/2016

Open Access 01.01.2016 | Review

Genetic Biomarkers of Barrett’s Esophagus Susceptibility and Progression to Dysplasia and Cancer: A Systematic Review and Meta-Analysis

verfasst von: John M. Findlay, Mark R. Middleton, Ian Tomlinson

Erschienen in: Digestive Diseases and Sciences | Ausgabe 1/2016

Abstract

Barrett’s esophagus (BE) is a common and important precursor lesion of esophageal adenocarcinoma (EAC). A third of patients with BE are asymptomatic, and our ability to predict the risk of progression of metaplasia to dysplasia and EAC (and therefore guide management) is limited. There is an urgent need for clinically useful biomarkers of susceptibility to both BE and risk of subsequent progression. This study aims to systematically identify, review, and meta-analyze genetic biomarkers reported to predict both. A systematic review of the PubMed and EMBASE databases was performed in May 2014. Study and evidence quality were appraised using the revised American Society of Clinical Oncology guidelines, and modified Recommendations for Tumor Marker Scores. Meta-analysis was performed for all markers assessed by more than one study. A total of 251 full-text articles were reviewed; 52 were included. A total of 33 germline markers of susceptibility were identified (level of evidence II–III); 17 were included. Five somatic markers of progression were identified; meta-analysis demonstrated significant associations for chromosomal instability (level of evidence II). One somatic marker of progression/relapse following photodynamic therapy was identified. However, a number of failings of methodology and reporting were identified. This is the first systematic review and meta-analysis to evaluate genetic biomarkers of BE susceptibility and risk of progression. While a number of limitations of study quality temper the utility of those markers identified, some—in particular, those identified by genome-wide association studies, and chromosomal instability for progression—appear plausible, although robust validation is required.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s10620-015-3884-5) contains supplementary material, which is available to authorized users.
An erratum to this article can be found at http://​dx.​doi.​org/​10.​1007/​s10620-016-4192-4.

Introduction

The importance of Barrett’s esophagus (BE) lies in its increasing prevalence and strong predisposition to esophageal adenocarcinoma (EAC) [1]. Western prevalence is estimated at 0.5–2.0 % [2] (a third of whom are asymptomatic [3]), conferring a 30- to 125-fold increased risk of developing EAC [4]. As EAC becomes more common [5], the benefits of predicting susceptibility to BE and detecting established metaplasia as soon as possible are numerous. Population screening overall is not presently feasible, but identifying both at-risk individuals and established metaplasia sooner provides windows for risk factor modification, chemoprevention, ablation, resection, and surveillance.
However, the natural history of BE is incompletely understood. Overall, the incidence of progression from metaplasia to high-grade dysplasia (HGD) or EAC is approximately 0.26–0.63 % per year [6], and that to EAC alone 0.12 % [7]. However, only a minority of patients progress to low-grade dysplasia (LGD), and of those that do similarly only a minority ultimately progress to HGD or EAC [8]. These progressive grades of dysplasia can help identify patients at particular risk (in particular, those with HGD in whom the risk is considerable) [9], but are insufficient in isolation.
There is therefore an immediate clinical need for biomarkers to predict both susceptibility to BE and progression. A major subtype is genetic variants, both germline and somatic. The potential of the latter, in particular, has been highlighted by a number of recent next-generation sequencing (NGS) studies of both EAC and BE, which have identified a number of candidate genes for further study [10]. However, no systematic reviews have been performed. The aims of this study were firstly to identify and evaluate all genetic markers tested in association with BE susceptibility and progression. Secondly, we aimed to identify markers with statistically significant associations and perform meta-analysis for those assessed by more than one study.

Methods

Inclusion Criteria

Studies testing associations between DNA markers [germline single nucleotide polymorphisms [SNP], somatic single nucleotide variants, insertions/deletions, copy number variants (CNV), loss of heterozygosity (LOH), microsatellite instability (MSI) or chromosomal instability (CIN)] and diagnosis/progression of BE were eligible; diagnosis was defined as endoscopic and histopathological evidence with or without intestinal metaplasia (IM) [13] and progression as histopathological progression from metaplasia to LGD/HGD/EAC or LGD to HGD/EAC, or HGD to EAC during surveillance endoscopies.

Exclusion Criteria

Studies comparing grades of metaplasia/dysplasia/malignancy within samples at one time point were excluded, as were studies comparing grades between patients. Studies using cell line, epigenetic, or expression data were excluded unless patient or DNA-specific data were available.
A search was performed in May 2014 of the PubMed and EMBASE databases, using the MOOSE and PRISMA guidelines [14] and the following term: [((((esophageal OR esophagus OR gastro esophageal)) AND (Barrett’s OR metaplasia OR columnar)) AND (genomic OR genetic OR genome OR pharmacogenetic OR pharmacogenomic OR amplification OR copy OR mutation OR polymorphism OR polymorphic OR variant OR deletion OR insertion OR locus OR loci OR allele OR ploidy OR instability OR biomarker))]. The bibliographies of retrieved articles were also searched.

Study Data

The following data were extracted: study methodology; variants and genes assessed; endpoints; population; and effect size [odds ratio (OR) or hazard ratio (HR)] and variance [standard error or confidence interval (CI)]. For studies not presenting OR, these were calculated using provided allele/variant frequencies. If reference SNP identification numbers were not provided (http://​www.​ncbi.​nlm.​nih.​gov/​dbSNP), these were mapped by searching cited methodology and performing in vitro polymerase chain reaction (http://​genome.​ucsc.​edu), with nucleotide flank BLAST® (http://​blast.​ncbi.​nlm.​nih.​gov).

Evidence Quality

Overall study and evidence quality were evaluated using the revised American Society of Clinical Oncology (ASCO) level of evidence (LOE) scale for biomarker research [15]. This stratifies study quality on the basis of trial design, patients and data, specimen collection, processing and archival, and statistical design and analysis from A to D (supplementary table 1), and uses this in conjunction with subsequent validation to stratify overall LOE for a marker from I to V (supplementary table 2). Methodological quality was appraised using the recommendations for tumor marker score (REMARK) guidelines modified by the authors, which scored study methodology in detail to generate a score from 0 to 17 (GWAS) and 0 to 18 (candidate studies; supplementary table 3). Reported associations were appraised for appropriate correction for multiple comparisons (Bonferroni method, false discovery rate, or multivariate analysis of all markers). If not undertaken, this was performed via post hoc Bonferroni correction. Significance was taken at p < 5×10−8 for genome-wide association studies (GWAS). Genomic quality criteria included reporting of genotyping call rate or providing data to allow its calculation, and assessment of Hardy–Weinberg equilibrium for germline variants.

Meta-Analysis

Meta-analysis was undertaken for markers assessed by more than one study, irrespective of correction for multiple comparisons (other than variants assessed by GWAS and non-GWAS due to major methodological differences) using RevMan v5.2 (Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration). Sensitivity analyses were performed on the basis of IM-only versus non-IM studies. In the case of studies reporting updated results using cohorts previously reported (i.e., population overlap), the most recent study was used.

Heterogeneity and Bias

Heterogeneity was estimated using I 2 and Chi-square statistics. For moderate heterogeneity (I 2 ≥ 50 %), a random-effects model was used. Funnel plots were reviewed for analyses of ≥5 studies [16]. Statistical assessment of these was not performed due to the low number of studies per variant. Publication bias was corrected using the “trim and fill” method [17].

Results

A total of 1516 articles were identified, of which 218 were duplicates. Therefore, 1298 articles were appraised on the basis of their title and abstract. Then, 251 full-text articles were retrieved, of which 52 met the inclusion criteria (Fig. 1).

Studies Assessing Germline Markers of Susceptibility

A total of 32 studies were included: 2 GWAS, 29 candidate studies, and 1 meta-analysis of 4 candidate studies (supplementary tables 4 and 5). All original studies were LOE C (indicating prospective observational registries, without standardized treatment and follow-up), published between 1999 and 2014. Median modified REMARK scores were 15/17 (GWAS) and 13/18 (candidate studies; range 7–16; supplementary table 6). A number of frequent methodological issues were identified. The most common of these included failure to blind investigators (n= 18; 58.1 %), perform/report quality control procedures (19; 61.3 %), and match cases and controls (23; 74.2 %). Similarly, a number of recurrent reporting issues were identified. The most common of these were comparing markers with established risk factors (30; 96.8 %), adjusting for multiple comparisons (22; 71.0 %) and confounding factors (20; 64.5 %), reporting power calculations (15; 48.4 %) and reporting multivariate effect sizes (30; 96.8 %). Of the 27 studies assessing multiple variants, 24 did not perform multivariate analysis (88.9 %). For GWAS, there were a mean 5507 cases and 14,159 controls. For candidate studies, 134 cases and 196 controls. There were a number of cases of study population overlap, with 13 studies reported on subjects drawn from one of three populations.

Variants Associated with BE Susceptibility

A total of 187 candidate variants/haplotypes were tested. Twenty-eight significant associations were reported, of which 16 were excluded (Table 2). Twelve were therefore associated with BE (Table 1). However, for 2 of these (rs6785049 and rs9344) precise p values were not provided to allow for Bonferroni correction.
Table 1
Reported germline markers of Barrett’s esophagus susceptibility
Marker
Individual studies
Association (meta-analysis/individual study)
LOE
Variant
Gene
Association
EV
LOE
Ethnicity
Methodology
Variant
Wild-type
None
Phenotype and OR (95 % CI)
I 2
Chi
p
n
III
rs9257809
MHC region
Su et al. [32], Palles et al. [33]
  
A
C
Caucasian
Multistage GWAS
Pro/retro archived
BEA 1.21 (1.13–1.28)
NA
NA
4.09 × 10−9
1
 
rs9936833
FOXF1
   
C
   
BEA 1.14 (1.10–1.19)
  
2.74 × 10−10
 
 
rs3072
GDF7
   
A
   
BEA 1.14 (1.09–1.18)
  
1.75 × 10−11
 
 
rs2701108
TBX5
   
A
   
BEA 0.90 (0.86–0.93)
  
7.48 × 10−9
 
 
rs2687201
FOXP1
   
T
   
BEA 1.16 (1.10–1.23)
  
4.61 × 10−8
 
III
rs3776082
CDX1
Ren et al. [43]
  
AA
C
Caucasian
Candidate
Prospective
BE-v-RE 4.02 (1.80–9.00)
NA
NA
<0.005
1
III
rs2237091
CDX1
 
Ren et al. [43]
 
AA
C
Caucasian
Candidate
Prospective
BE-v-RE 0.28 (0.13–0.59)
NA
NA
<0.005
1
III
rs717746
CDX1
Ren et al. [43]
  
GG
C
Caucasian
Candidate
Prospective
BE-v-RE 3.65 (1.73–7.69
NA
NA
<0.005
1
III
rs4444903
EGFR
Menke et al. [20]
  
GG
C
Caucasian*
Candidate
Retro archived
BEA 3.00 (1.50–6.20)
NA
NA
0.002
1
III
rs917997
IL18RAP
 
Babar et al. [29]
 
CC
C
Caucasian**
Candidate
Retro archived
BEA 0.59 (0.43–0.80)
NA
NA
6.00 × 10−4
1
III
rs6785049
PXR
van de Winkel et al. [44]
  
G
C
Caucasian*
Candidate
Retro archived
BE 1.36 (1.03–1.79)
NA
NA
<0.05 (NS)
1
III
rs6214
IGF1
 
McElholm [21]
 
AA
C
Caucasian**
Candidate
Retro archived
BEA 0.43 (0.24–0.75)
NA
NA
3.00 × 10−3
1
III
rs2229765 + BMI > 30
IGF1R
MacDonald et al. [22]
  
A
C
Caucasian
Candidate
Retro archived
BEA 3.11 (1.12–8.63)
NA
NA
<0.05 (NS)
1
III
rs3212227
IL12B
Moons et al. [26]
  
C
C
Caucasian*
Candidate
Retro archived
BEA 1.82 (1.17–2.69)
NA
NA
7.00 × 10−3
1
II
rs1695
GSTP
Bull et al. [18]
  
G
C
Caucasian
Meta-analysis
BE 1.50 (1.16–1.95)
NA
NA
<0.01
4
Kala et al. [19]
  
G
 
Caucasian
Candidate (pro)
  
Murphy et al. [45]
G
 
Caucasian
Candidate (retro)
  
Casson et al. [46]
G
 
NP
Candidate (pro)
  
van Lieshout et al. [47]
G
 
Caucasian
Candidate (pro)
III
rs9344
CCND1
Casson et al. [31]
  
AA
C
NP
Candidate (pro)
BEA 3.69 (1.46–9.29)
NA
NA
<0.05 (NS)
1
III
rs25487
XRCC1
Casson et al. [48]
 
Ferguson et al. [49]
AA
C
NP
Candidate (pro)
BEA 0.65 (0.25–1.68)
71 %
3.42
0.370
2
AA
C
Caucasian
Candidate (retro)
LOE level of evidence, EV effect variant, NS nonsignificant, NP not presented, BE Barrett’s esophagus, A adjusted OR, OR odds ratio, CI confidence interval, pro prospective, retro retrospective, RE reflux esophagitis, NA not applicable
*,** denote overlapping populations
Table 2
Reported germline markers of Barrett’s esophagus excluded on the basis of multiple comparisons
Marker
Individual studies
Association (meta-analysis/individual study)
LOE
Variant
Gene
Association
EV
LOE
Ethnicity
Methodology
Variant
Wild-type
Phenotype and OR (95 % CI)
I 2
Chi
p
n
III
rs3776083
CDX1
Ren et al. [43]
 
AG
C
Caucasian
Candidate pro
BE-v-RE 1.91 (1.09–3.33)
NA
NA
0.02
1
III
rs4769585
CDX2
Ren et al. [43]
 
CT
C
Caucasian
Candidate pro
BE-v-RE 2.68 (1.20–5.98)
NA
NA
0.02
1
III
rs3812863
CDX2
Ren et al. [43]
 
AG
C
Caucasian
Candidate pro
BE-v-RE 2.53 (1.24–5.14)
NA
NA
0.01
1
III
rs1143634
IL1B
 
Izakovicoka-Holla et al. [27]
T
 
Caucasian (Czeck)
Candidate pro
BE -v-RE 0.56 (0.33–0.93)
NA
NA
0.016
1
III
VNTR polymorphism
IL1RN
 
Izakovicoka-Holla et al. [27]
1/2
 
Caucasian (Czeck)
Candidate pro
BE 0.62 (0.42–0.94)
NA
NA
0.015
1
III
rs1800587/rs16944/
rs1143634/IL1RN
IL1B/ILRN
 
Izakovicoka-Holla et al. [27]
C/T/C/long
 
Caucasian (Czeck)
Candidate pro
BE 1.95 (1.09–1.30)
NA
NA
0.03
1
III
rs1800587/rs16944/r
s1143634/IL1RN
IL1B/ILRN
 
Izakovicoka-Holla et al. [27]
T/C/C/long
 
Caucasian (Czeck)
Candidate pro
BE 2.67 (1.15–6.24)
NA
NA
0.02
1
III
rs909253
TNFB
Menke et al. [50]
 
AA
C
Caucasian (Netherlands)
Candidate retro archived
BEA 1.98 (1.03–3.81)
NA
NA
0.04
1
III
rs2305764
Myo9b
Menke et al. [51]
 
GG
C
Caucasian (Netherlands)
Candidate retro archived
BEA 2.96 (1.10–7.99)
NA
NA
0.032
1
III
rs1946518
IL18
Babar et al. [29]
 
CC
 
Caucasian (Irish)
Candidate retro archived
BEA 1.45 (1.07–1.98)
NA
NA
0.02
1
III
rs1799750
MMP1
Bradbury et al. [52]
 
GG
 
Caucasian (USA)
Candidate retro archived
BEA 2.21 (1.08–4.53)
NA
NA
0.03
1
III
rs11209026
IL23R
Gaj et al. [30]
 
A
 
Caucasians (Polish)
Candidate pro
BE 3.49 (1.37–8.90)
NA
NA
0.011
1
III
rs1800566
NQO1
di Martino et al. [53]
 
T
 
NP (UK)
Candidate pro
BEA-v-RE 0.22 (0.07–0.76)
NA
NA
0.016
1
III
B7
HLA
Rajendra [54]
 
B7
 
Asian (India)
Candidate pro
BEEx 21.4 (1.22–372
NA
NA
0.036
1
III
rs415958/rs16944
IL1RA/IL1B
Gough et al. [28]
 
CC/A
 
NS (UK)
Candidate pro
BE-v-RE 9.5 (1.19–75.9)
NA
NA
0.010
1
III
rs1800896
IL10
Gough et al. [28]
 
CC
 
NS (UK)
Candidate pro
BE-v-RE 1.84 (1.04–3.28)
NA
NA
0.035
1
LOE level of evidence, EV effect variant, NP not presented; BE = Barrett’s esophagus; A adjusted OR, OR odds ratio, CI confidence interval, pro prospective, retro retrospective, NA not applicable, RE reflux esophagitis, Ex extrapolated
The rs1695 (GSTP1) was assessed by 4 studies, which underwent meta-analysis by Bull et al. [18]. This calculated an OR of 1.50 (95 % CI 1.16–1.95 p = not presented; LOE II). While derived from a large total cohort (434 cases and 738 controls), none of the four studies adjusted for risk factors; indeed the sole study finding a significant association had only 22 cases [19]. The GSTM1 null genotype was also assessed by 4 studies and underwent updated meta-analysis in this study. Overall, no association was demonstrated. On sensitivity analysis, a significant negative association was apparent for the 2 studies not requiring IM [17, 20], although the relevance of this is unclear. Meta-analysis was performed for 5 other variants, none of which demonstrated associations (Table 3).
Table 3
Meta-analyzed markers assessed in association with BE susceptibility
Marker
Individual study
Meta-analysis
Association
Variant
Gene
EV
Variant
Wild-type
No association
OR (95 % CI)
I 2
Chi
p
n
Null
GSTM1
Null
Null
 
Kadioglu et al. [55]
BE 0.83 (0.61–1.12)
27 %
4.12
0.220
4
     
Kala et al. [19]
BEno IM 0.66 (0.44–0.99)
0 %
0.64
0.420
2
     
Casson et al. [46]
BEIM 1.12 (0.70–1.80)
0 %
0.64
0.630
2
     
van Lieshout et al. [56]
     
rs4880
SOD2
T
T
 
Kadioglu et al. [55],
di Martino et al. [49]
BEIM 0.90 (0.65–1.24)
0.00
0.07
0.520
3
     
Murphy et al. [45]
     
rs16944
IL1B
AA
AA
 
Izakovicova-Holla et al. [27],
Gough et al. [28]
BE 1.13 (0.63–2.03)
0.00
0.00
0.680
2
rs1052133
OGG1
G
G
 
Kadioglu et al. [55],
Ferguson et al. [49]
BEA IM 1.34 (0.59–3.09)
67 %
3.01
0.480
2
rs25487
XRCC1
TT
TT
Casson et al. [48]
Ferguson et al. [49]
BEA IM 0.65 (0.25–1.68)
71 %
3.42
0.370
2
rs13181
ERCC2
CC
CC
 
Casson et al. [48],
Fergusons et al. [49]
BEA IM 0.92 (0.57–1.48)
31 %
1.44
0.730
2
EV effect variant, OR odds ratio, CI confidence interval, IM intestinal metaplasia, BE Barrett’s esophagus
Of the 12 significant candidate associations reported, only rs1695 (GSTP1) and rs25487 (XRCC1) were assessed by more than one study. This notwithstanding, 5 appear relatively robust on the basis of adjustment for clinical covariates. These include 3 growth factor variants: rs444903 (EGFR [20]; notably associated with reflux esophagitis and EAC), rs6214 (IGF1 [21]), and rs2229765 (IGF1R) [22]. Two interleukin variants also appear plausible: rs3212227 (IL12B) and rs917997 (IL18RAP) [2325], with the former demonstrated to be independent of all other tested genotypes [26]. A number of other associations were reported in the IL1 [27, 28], IL10 [28], IL18 [29], and IL23 [30] clusters. Of these, however, only wild-type rs917997 (IL18RAP) persisted following correction for multiple comparisons [29].
The remaining 5 candidate variants included associations with 3 caudal homeobox 1 (CDX1) variants: rs3776082, rs2237091, and rs717767. The authors demonstrated these variants to be significantly associated with established risk factors for BE: age, gender, and the presence of hiatus hernia. However, multivariate analysis was not performed to demonstrate whether the association of these variants with BE was independent of these. Of the remaining 2, an association was demonstrated for the rs6785049 (NR1I2) variant; however, similarly this was not adjusted for risk factors. This was performed for the rs9344 (CCND1 [31]) variant, although the p value was not published.
Of the 5 GWAS variants identified, 4 were identified by the Wellcome Trust Centre Case Control Consortium (WTCCC). Two were identified by the initial report [32]: rs9257809 within the major histocompatibility complex (MHC; OR 1.21 [1.13–1.28]; p = 4.09 × 10−9) and rs9936833 (related to FOXF1; OR 1.14 [1.10–1.19]; p = 2.74 × 10−10). Subsequent replication identified rs3072 (related to GDF7: OR 1.14 [1.091.18]; p = 1.80 × 10−11) and rs2701108 (related to TBX5: OR 0.90 [0.86–0.93]; p = 7.50 × 10−9) [33]. All remained significant when meta-analyzed with data from the Barrett’s and Esophageal Adenocarcinoma Consortium (BEACON) GWAS. The first 2 were also associated with EAC in the BEACON dataset and an independent candidate study [34]. Mechanistically, GWAS arrays rely on linkage disequilibrium between SNPs, by which SNPs act as proxies for others (i.e., genotyping one SNP allows the genotypes of others to be inferred with confidence). This “tagging”, however, means that variants identified may be functional or may in fact be bystanders tagging other SNPs [35]. However, the loci overall and related genes are consistent with the roles of immune-mediated inflammation (MHC) and thoracoembryogenesis (FOXF1, GDF7 and TBX5) in BE.
The BEACON GWAS identified 3 loci associated with either BE or EAC, although none reached the genome-wide threshold for BE alone. However, on subsequent meta-analysis of both GWAS, rs2687201 (FOXP1, similarly involved in developmental regulation) was significantly associated with BE alone, in addition to a further variant associated with either BE/EAC [33].

Studies Assessing Germline Variants Associated with Progression

One study was identified (supplementary table 7). LOE was C; modified REMARK score was 13.5/19. This assessed 4 variants in the IGF axis; none were associated with progression.

Studies Assessing Somatic Variants Associated with Progression

Sixteen studies were identified, published between 1989 and 2012 (supplementary table 8). LOE was C for 12 and D for 4. Mean modified REMARK score was 13.2 (range 10.5–18.5/19; supplementary table 9). Five classified progression as HGD/EAC, 10 as EAC, and 1 as EAC/CIN. Again, a number of recurrent methodological issues were identified. These included failure to: blind investigators (12; 75.0 %), perform appropriate quality control/reproducibility (9; 56.3 %), match controls (14; 87.5 %), and appropriate power calculations (14; 87.5 %). Recurrent reporting issues included failure to: report univariate association effects (9; 60.0 %), adjust for risk factors (particularly the presence of dysplasia at baseline; 14; 87.5 %), and fully report coefficients of multivariate models (13; 81.3 %).

Variants Associated with Progression

Of 7 variants assessed, 5 associations were identified: CIN, CNV (>70 Mbp), TP53 LOH, p16 LOH, and mutant TP53 (Table 4). Meta-analysis was possible for CIN, which was assessed by 11 studies. These defined CIN variably as aneuploidy (4), tetraploidy (1), and aneuploidy/tetraploidy (6). Six of these studies were derived from independent sample archives. However, this was not clear for 4. All studies reported associations of CIN with progression. However, only two adjusted for confounding variables, including the presence of dysplasia [36, 37]. Another two, while not adjusting for dysplasia, did adjust for length of Barrett’s segment [38, 39], with CIN remaining significant.
Table 4
Reported somatic markers of Barrett’s esophagus progression to dysplasia/adenocarcinoma
Marker
Individual study
Association (meta-analysis/individual study)
LOE
Variant
Gene
Association
EV
LOE
Ethnicity
Phenotype
Adjusted for confounders?
Variant
None
Phenotype and OR/HR (95 % CI)
I 2
Chi
p
n
II
CIN
NA
Bird-Lieberman et al. [36]
 
CIN
C
NP
HGD/EAC
Yes (plus length, dysplasia)
HGD/EAC OR(A) IM PB 5.98 (2.10–17.1)
57 %
16.2
8.00 × 10−4
5
   
Sikkema et al. [37]
 
CIN
C
Cauc
HGD/EAC
Yes (plus dysplasia)
HGD/EAC HR(A) IM 1.36 (1.26–1.47)
0 %
0.98
<1.00 × 10−5
2
   
Chao et al. [57]*
 
CIN
C
NP
EAC
No
     
   
Galipeau et al. [38]*
 
CIN
C
NP
EAC
Yes (length, not dysplasia)
PB-adjusted for publication bias Fang 2004, Rabinovitch 2001,
    
   
Fang et al. [58]
 
CIN
C
NP
EAC
No
    
   
Maley et al. [39]*
 
CIN
C
NP
EAC
Yes (length, not dysplasia)
Teodori 1999
    
   
Rabinovitch et al. [59]*
 
CIN
C
NP
EAC
Yes (length, not dysplasia)
     
   
Reid et al. [9]**
 
CIN
D
NP
EAC
No
Overlapping populations***
    
   
Teodori et al. [60]
 
CIN
C
NP
HGD/EAC
No
     
   
Reid et al. [8]**
 
CIN
C
NP
EAC
No
     
   
James et al. [61]
 
CIN
D
NP
EAC
No
     
III
CNV
NA
Paulson et al. [62]
 
>70 Mbp
C
NP
EAC/CIN
No
EAC/CIN HR 4.90 (1.60–14.8)
NA
NA
4.70 × 10−3 (NS)
1
III
LOH
TP53
Galipeau et al. [38]*
 
LOH
C
NP
EAC
Yes (length, not dysplasia)
EAC HRA 5.4 (2.5–12.0)
NA
NA
<1 × 10−3
1
   
Maley et al. [39]*
 
LOH
C
        
   
Reid et al. [63]*
 
LOH
C
        
III
LOH
P16
Galipeau et al. [38]*
Maley et al. [39]*
LOH
C
NP
EAC
Yes (length, not dysplasia)
EACA HR 2.4 (1.0–5.5)
NA
NA
0.045
1
     
LOH
C
NP
EAC
Yes (length, not dysplasia)
     
III
Mutant
TP53
 
Galipeau et al. [38]*
Mutant
C
NP
EAC
No
EAC HRA 1.27 (1.07–1.50)
NA
NA
<0.01 (NS)
1
   
Maley et al. [39]*
 
Mutant
C
NP
EAC
Yes (length, not dysplasia)
     
   
Dolan et al. [64]
 
Mutant
C
NP
EAC
No
No meta possible as study population overlap and differing effect measures
    
LOE level of evidence, EV effect variant, OR odds ratio, HR hazard ratio, CI confidence interval, CIN chromosomal instability, NA not applicable, NP not presented, NS nonsignificant, HGD high-grade dysplasia, EAC esophageal adenocarcinoma, A adjusted
*,**,*** denote overlapping populations
Meta-analysis was performed for both OR and HR of progression to HGD/EAC. Significant associations were demonstrated for both (Table 4; Fig. 2). All studies included patients with IM only. Meta-analyzed OR was 5.98 (2.10–17.1; p = 8.00 × 10−4; n = 5 studies; following exclusion of overlapping studies and correction for publication bias). However, only one of these studies adjusted for the presence of dysplasia. Meta-analyzed HR was 1.36 (1.26–1.47; n = 2 studies; p < 1.00 × 10−5; following exclusion of one overlapping study).Overall LOE for CIN was II. While more than one study assessed mutant TP53 (n = 3) and LOH TP53 (n = 2), meta-analysis was not possible for either. Two studies for each were derived from the same populations, and the third TP53 study used a different measure of effect size.

Variants Associated with Risk of Progression Following Photodynamic Therapy

Three studies were identified (supplementary table 10). LOE was C for 2 and D for 1. Mean modified REMARK score was 16.2/20 (range 14.5–19; supplementary table 7). For 2 studies, while endoscopic mucosal resection was variably performed, this was only controlled for in 1 study. A total of 6 variants were assessed; while 5 were assessed by 2 studies, meta-analysis could not be performed due to population overlap. CIN at both 4 and 12 months was reported to be associated with risk of progression (Table 5).
Table 5
Reported somatic biomarkers of Barrett’s esophagus progression to dysplasia/adenocarcinoma following photodynamic therapy
Marker
Individual study
Association (meta-analysis/individual study)
LOE
Variant
Gene
Association
EV
LOE
Ethnicity
Phenotype
Adjusted for confounders?
Variant
None
Phenotype HR (95 % CI)
I 2
Chi
p
n
IV
CIN
NA
Dunn et al. [65]
 
CIN 4 m post-PDT
D
NS
HGD
Yes
EAC/HGD HRA 4.10 (1.30–13.0)
NA
NA
9.00 × 10−3
1
CIN 12 m post-PDT
EAC/HGD HRA 3.60 (1.05–12.3)
2.90 × 10−3
LOE level of evidence, CIN chromosomal instability, EV effect variant, HR hazard ratio, CI confidence interval, PDT photodynamic therapy, HGD high-grade dysplasia, EAC esophageal adenocarcinoma

Discussion

We believe this review to be the first to identify, synthesize, and evaluate the evidence for genetic markers of BE susceptibility and risk of progression. Thirty-three susceptibility markers were identified; however, just 17 remained significant after correcting for multiple comparisons. Five (rs9257809, rs9936833, and subsequently rs3072, rs2701108, and rs2687201) were derived from GWAS and are therefore most likely to be reproducible. Of the 6 candidate markers assessed by more than one study, meta-analysis was supportive for one (rs1695, GSTP1). Five non-meta-analyzed variants affecting either growth factors or inflammatory cytokines appear plausible and therefore represent priorities for validation: rs444903 (EGFR), rs6214 (IGF1), rs2229765 (IGF1R), rs3212227 (IL12B), and rs917997 (IL18RAP).
No germline markers of progression risk were identified. However, 5 somatic markers were reported, plus another of progression following photodynamic therapy. Meta-analysis was possible for CIN, demonstrating significant effects for both HR and OR after correction for publication bias. However, there was considerable heterogeneity regarding definition of CIN, duration, and frequency of follow-up, confounding risk factors (e.g., prevalence of HGD) in addition to minimum follow-up periods. Notably, however, both studies adjusting for HGD [36, 37] did demonstrated convincing effects.
The robustness of the associations between CIN and progression suggests it to be of immediate clinical utility. CIN is a constituent of genomic instability, a state of erroneous progression through the cell cycle. Inaccurate DNA replication, repair, and chromosomal segregation, results in accumulation of genomic errors and is a major factor driving tumorigenesis. CIN is associated with worse stage and prognosis in a range of tumors including esophageal [40] and has been demonstrated in a quarter of patients with BE [36]. Importantly, this subset appears to be at significantly higher risk of malignant progression, which can be readily demonstrated by flow cytometry. This does, however, remain somewhat imprecise and is unable to distinguish between stable and unstable, simple and complex abnormalities. Despite this, the incorporation of CIN into a biomarker panel such as that reported by Bird-Lieberman et al. [36] (comprising age, CIN, dysplasia, TP53 and Cyclin A expression, sialyl Lewis antigens, Aspergillus oryzae lectin, and binding of wheat germ agglutinin) may provide invaluable information with which to personalize management of BE.
By contrast, the immediate benefits of germline susceptibility biomarkers are less tangible. As predicted by the “common disease-common variant” hypothesis of complex traits, both GWAS suggested many common variants of small effect to contribute to development of BE. Consequently, germline associations may be weaker and more complex. Whilst this gives the potential to identify novel biology, variants may have little utility in isolation.
A number of recurrent methodological issues were identified, limiting the generalizability of reported variants. Ultimately, validation studies for prioritized variants should be designed with these in mind, with particular emphasis on the interaction between genomic and clinical factors. Other issues to be addressed include disparity as diagnostic criteria; confirmation of IM is a prerequisite in the USA, yet is not required in the UK [41], although this did not alter the findings of this review.
A number of exploratory NGS studies have recently been performed for both BE and EAC. These have served to highlight the mutational complexity of both conditions, providing biological context for markers and their genes (which are often considered in isolation) as well as suggesting new variants and genes for study. Recently, Streppel et al. [11] performed whole genome sequencing (WGS) of one patient, comparing normal squamous epithelium with metaplastic and neoplastic epithelium. This identified somatic nonsense mutations in genes including AT-rich interactive domain 1A (ARID1A), a member of the SWI/SNF family involved in gene expression via chromatin remodeling, which has been independently identified as a driver gene of EAC by of other NGS studies. The authors found ARID1A loss of expression to become progressively more common during the metaplasia-dysplasia-adenocarcinoma sequence, and to be associated with aberrant cellular proliferation and invasion in a knock-down model. NGS studies also provide valuable contextual information as to mutational spectra, as well as clonal and linear evolution to better understand to development of somatic mutations and genomic instability. Recently, Weaver et al. [12] performed WGS of 112 EACs, similarly identifying a number of significantly and recurrently mutated genes. One hundred and seven BE samples were then genotyped, with the notable finding that most such mutations were already present in non-dysplastic epithelium; just TP53 and SMAD4 mutations occurred later, in HGD and EAC. The advent of third-generation sequencing, for example from single cells, will undoubtedly shed yet further light on this process. Additionally, germline rather than somatic mutations in EAC driver genes have been shown to predispose to EAC, although this has yet to be demonstrated in BE [42].
This review has a number of limitations. While we searched two databases using a comprehensive search term, it is possible that relevant publications (including non-English articles) were not identified. As discussed, meta-analysis for CIN was performed within the context of considerably heterogeneity and must be interpreted with caution. Only one meta-analysis (CIN and OR of progression) involved more than 5 studies, with considerable funnel plot asymmetry, largely due to small studies reporting large effect sizes with significant variance. This was interpreted as publication bias, and was adjusted, without altering statistical significance. In addition, the “trim and fill” method used to correct for possible bias while widely used does make assumptions regarding the necessity for plot symmetry, while not incorporating study methodology. Unfortunately, the limited number of studies prevented useful meta-regression to assess this further. There are also limitations in using the revised ASCO guidelines, as these do not fully represent the complexity of methodological quality and also disagreement between studies. We therefore used modified REMARK guidelines to provide a further level of criticism. In particular, the ASCO guidelines do not allow for differences in methodology (for example, GWAS versus candidate studies). And more generically, while genomic biomarkers are typically considered in isolation, in reality their utility depends on innumerable variables (including transcriptional, translational and proteomic regulation, and clinical and environmental factors). Consequently, establishing their true utility will require parallel processing and consideration of these contexts.
In conclusion, this review has identified, evaluated, and synthesized the evidence for genomic biomarkers of BE susceptibility and dysplastic/malignant progression. Seventeen germline markers of susceptibility, 5 somatic markers of progression, and 1 marker of relapse following photodynamic therapy were identified. Meta-analysis demonstrated CIN to be a particularly plausible and clinically useful marker of progression and one which can be demonstrated readily. However, the overall evidence base is characterized by widespread methodological issues, which limit the immediate clinical utility of these markers. Consequently, larger studies with more robust design are required to validate these markers, identify novel variants, and incorporate them into clinical practice.

Acknowledgments

Core funding is provided to the Wellcome Trust Centre for Human Genetics from the Wellcome Trust (090532/Z/09/Z).

Compliance with ethical standards

Conflict of interest

Mark R. Middleton has received payment for advisory/consulting roles within the last 2 years from Amgen, Bristol-Myers Squibb, GlaxoSmithKline, Merck, Millennium, and Roche, and institutional funding from Amgen, AstraZeneca, Bristol-Myers Squibb, Clovis, Eisai, GlaxoSmithKline, Immunocore, Johnson & Johnson, Merck, Millennium, Novartis, Pfizer, Roche and Vertex. The authors declare no conflicts of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License (http://creativecommons.org/licenses/by-nc/4.0/), which permits any noncommercial use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Barrett NR. Chronic peptic ulcer of the oesophagus and ‘oesophagitis’. Br J Surg. 1950;38:175–182.CrossRefPubMed Barrett NR. Chronic peptic ulcer of the oesophagus and ‘oesophagitis’. Br J Surg. 1950;38:175–182.CrossRefPubMed
2.
Zurück zum Zitat Cameron AJ, Lomboy CT. Barrett’s esophagus: age, prevalence, and extent of columnar epithelium. Gastroenterology. 1992;103:1241–1245.PubMed Cameron AJ, Lomboy CT. Barrett’s esophagus: age, prevalence, and extent of columnar epithelium. Gastroenterology. 1992;103:1241–1245.PubMed
3.
Zurück zum Zitat Gerson LB, Edson R, Lavori PW, Triadafilopoulos G. Use of a simple symptom questionnaire to predict Barrett’s esophagus in patients with symptoms of gastroesophageal reflux. Am J Gastroenterol. 2001;96:2005–2012.CrossRefPubMed Gerson LB, Edson R, Lavori PW, Triadafilopoulos G. Use of a simple symptom questionnaire to predict Barrett’s esophagus in patients with symptoms of gastroesophageal reflux. Am J Gastroenterol. 2001;96:2005–2012.CrossRefPubMed
4.
Zurück zum Zitat Wild CP, Hardie LJ. Reflux, Barrett’s oesophagus and adenocarcinoma: burning questions. Nat Rev Cancer. 2003;3:676–684.CrossRefPubMed Wild CP, Hardie LJ. Reflux, Barrett’s oesophagus and adenocarcinoma: burning questions. Nat Rev Cancer. 2003;3:676–684.CrossRefPubMed
5.
Zurück zum Zitat Lepage C, Rachet B, Jooste V, Faivre J, Coleman MP. Continuing rapid increase in esophageal adenocarcinoma in England and Wales. Am J Gastroenterol. 2008;103:2694–2699.CrossRefPubMed Lepage C, Rachet B, Jooste V, Faivre J, Coleman MP. Continuing rapid increase in esophageal adenocarcinoma in England and Wales. Am J Gastroenterol. 2008;103:2694–2699.CrossRefPubMed
6.
7.
Zurück zum Zitat Hvid-Jensen F, Pedersen L, Drewes AM, Sorensen HT, Funch-Jensen P. Incidence of adenocarcinoma among patients with Barrett’s esophagus. N Engl J Med. 2011;365:1375–1383.CrossRefPubMed Hvid-Jensen F, Pedersen L, Drewes AM, Sorensen HT, Funch-Jensen P. Incidence of adenocarcinoma among patients with Barrett’s esophagus. N Engl J Med. 2011;365:1375–1383.CrossRefPubMed
8.
Zurück zum Zitat Reid BJ, Blount PL, Rubin CE, Levine DS, Haggitt RC, Rabinovitch PS. Flow-cytometric and histological progression to malignancy in Barrett’s esophagus: prospective endoscopic surveillance of a cohort. Gastroenterology. 1992;102:1212–1219.PubMed Reid BJ, Blount PL, Rubin CE, Levine DS, Haggitt RC, Rabinovitch PS. Flow-cytometric and histological progression to malignancy in Barrett’s esophagus: prospective endoscopic surveillance of a cohort. Gastroenterology. 1992;102:1212–1219.PubMed
9.
Zurück zum Zitat Reid BJ, Levine DS, Longton G, Blount PL, Rabinovitch PS. Predictors of progression to cancer in Barrett’s esophagus: baseline histology and flow cytometry identify low- and high-risk patient subsets. Am J Gastroenterol. 2000;95:1669–1676.PubMedPubMedCentral Reid BJ, Levine DS, Longton G, Blount PL, Rabinovitch PS. Predictors of progression to cancer in Barrett’s esophagus: baseline histology and flow cytometry identify low- and high-risk patient subsets. Am J Gastroenterol. 2000;95:1669–1676.PubMedPubMedCentral
10.
Zurück zum Zitat Streppel MM, Lata S, DelaBastide M, et al. Next-generation sequencing of endoscopic biopsies identifies ARID1A as a tumor-suppressor gene in Barrett’s esophagus. Oncogene. 2014;33:347–357.CrossRefPubMedPubMedCentral Streppel MM, Lata S, DelaBastide M, et al. Next-generation sequencing of endoscopic biopsies identifies ARID1A as a tumor-suppressor gene in Barrett’s esophagus. Oncogene. 2014;33:347–357.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Dulak AM, Stojanov P, Peng S, et al. Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat Genet. 2013;45:478–486.CrossRefPubMedPubMedCentral Dulak AM, Stojanov P, Peng S, et al. Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat Genet. 2013;45:478–486.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Weaver JM, Ross-Innes CS, Shannon N, et al. Ordering of mutations in preinvasive disease stages of esophageal carcinogenesis. Nat Genet. 2014;46:837–843.CrossRefPubMedPubMedCentral Weaver JM, Ross-Innes CS, Shannon N, et al. Ordering of mutations in preinvasive disease stages of esophageal carcinogenesis. Nat Genet. 2014;46:837–843.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Hellier MD, Shepherd NA. BSG Guidelines in Gastroenterology: Diagnosis of Columnar-Lined Oesophagus. British Society of Gastroenterology; 2005:13–17. Hellier MD, Shepherd NA. BSG Guidelines in Gastroenterology: Diagnosis of Columnar-Lined Oesophagus. British Society of Gastroenterology; 2005:13–17.
14.
Zurück zum Zitat Stroup DF, Berlin JA, Morton SC, et al. Meta-analysis of observational studies in epidemiology: a proposal for reporting. Meta-analysis of Observational Studies in Epidemiology (MOOSE) group. JAMA. 2000;283:2008–2012.CrossRefPubMed Stroup DF, Berlin JA, Morton SC, et al. Meta-analysis of observational studies in epidemiology: a proposal for reporting. Meta-analysis of Observational Studies in Epidemiology (MOOSE) group. JAMA. 2000;283:2008–2012.CrossRefPubMed
15.
Zurück zum Zitat Simon RM, Paik S, Hayes DF. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst. 2009;101:1446–1452.CrossRefPubMedPubMedCentral Simon RM, Paik S, Hayes DF. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst. 2009;101:1446–1452.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Sterne JA, Sutton AJ, Ioannidis JP, et al. Recommendations for examining and interpreting funnel plot asymmetry in meta-analyses of randomised controlled trials. BMJ. 2011;343:d4002.CrossRefPubMed Sterne JA, Sutton AJ, Ioannidis JP, et al. Recommendations for examining and interpreting funnel plot asymmetry in meta-analyses of randomised controlled trials. BMJ. 2011;343:d4002.CrossRefPubMed
17.
Zurück zum Zitat Sutton AJ, Duval SJ, Tweedie RL, Abrams KR, Jones DR. Empirical assessment of effect of publication bias on meta-analyses. BMJ. 2000;320:1574–1577.CrossRefPubMedPubMedCentral Sutton AJ, Duval SJ, Tweedie RL, Abrams KR, Jones DR. Empirical assessment of effect of publication bias on meta-analyses. BMJ. 2000;320:1574–1577.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Bull LM, White DL, Bray M, Nurgalieva Z, El-Serag HB. Phase I and II enzyme polymorphisms as risk factors for Barrett’s esophagus and esophageal adenocarcinoma: a systematic review and meta-analysis. Dis Esophagus. 2009;22:571–587.CrossRefPubMedPubMedCentral Bull LM, White DL, Bray M, Nurgalieva Z, El-Serag HB. Phase I and II enzyme polymorphisms as risk factors for Barrett’s esophagus and esophageal adenocarcinoma: a systematic review and meta-analysis. Dis Esophagus. 2009;22:571–587.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Kala Z, Dolina J, Marek F, Izakovicova Holla L. Polymorphisms of glutathione S-transferase M1, T1 and P1 in patients with reflux esophagitis and Barrett’s esophagus. J Hum Genet. 2007;52:527–534.CrossRefPubMed Kala Z, Dolina J, Marek F, Izakovicova Holla L. Polymorphisms of glutathione S-transferase M1, T1 and P1 in patients with reflux esophagitis and Barrett’s esophagus. J Hum Genet. 2007;52:527–534.CrossRefPubMed
20.
Zurück zum Zitat Menke V, Pot RG, Moons LM, et al. Functional single-nucleotide polymorphism of epidermal growth factor is associated with the development of Barrett’s esophagus and esophageal adenocarcinoma. J Hum Genet. 2012;57:26–32.CrossRefPubMed Menke V, Pot RG, Moons LM, et al. Functional single-nucleotide polymorphism of epidermal growth factor is associated with the development of Barrett’s esophagus and esophageal adenocarcinoma. J Hum Genet. 2012;57:26–32.CrossRefPubMed
21.
Zurück zum Zitat McElholm AR, McKnight AJ, Patterson CC, et al. A population-based study of IGF axis polymorphisms and the esophageal inflammation, metaplasia, adenocarcinoma sequence. Gastroenterology. 2010;139:204-e3–212-e3.CrossRef McElholm AR, McKnight AJ, Patterson CC, et al. A population-based study of IGF axis polymorphisms and the esophageal inflammation, metaplasia, adenocarcinoma sequence. Gastroenterology. 2010;139:204-e3–212-e3.CrossRef
22.
Zurück zum Zitat MacDonald K, Porter GA, Guernsey DL, Zhao R, Casson AG. A polymorphic variant of the insulin-like growth factor type I receptor gene modifies risk of obesity for esophageal adenocarcinoma. Cancer Epidemiology. 2009;33:37–40.CrossRefPubMed MacDonald K, Porter GA, Guernsey DL, Zhao R, Casson AG. A polymorphic variant of the insulin-like growth factor type I receptor gene modifies risk of obesity for esophageal adenocarcinoma. Cancer Epidemiology. 2009;33:37–40.CrossRefPubMed
23.
Zurück zum Zitat O’Riordan JM, Abdel-latif MM, Ravi N, et al. Proinflammatory cytokine and nuclear factor kappa-B expression along the inflammation–metaplasia–dysplasia–adenocarcinoma sequence in the esophagus. Am J Gastroenterol. 2005;100:1257–1264.CrossRefPubMed O’Riordan JM, Abdel-latif MM, Ravi N, et al. Proinflammatory cytokine and nuclear factor kappa-B expression along the inflammation–metaplasia–dysplasia–adenocarcinoma sequence in the esophagus. Am J Gastroenterol. 2005;100:1257–1264.CrossRefPubMed
24.
Zurück zum Zitat Jenkins GJ, Harries K, Doak SH, et al. The bile acid deoxycholic acid (DCA) at neutral pH activates NF-kappaB and induces IL-8 expression in oesophageal cells in vitro. Carcinogenesis. 2004;25:317–323.CrossRefPubMed Jenkins GJ, Harries K, Doak SH, et al. The bile acid deoxycholic acid (DCA) at neutral pH activates NF-kappaB and induces IL-8 expression in oesophageal cells in vitro. Carcinogenesis. 2004;25:317–323.CrossRefPubMed
25.
Zurück zum Zitat Fitzgerald RC, Abdalla S, Onwuegbusi BA, et al. Inflammatory gradient in Barrett’s oesophagus: implications for disease complications. Gut. 2002;51:316–322.CrossRefPubMedPubMedCentral Fitzgerald RC, Abdalla S, Onwuegbusi BA, et al. Inflammatory gradient in Barrett’s oesophagus: implications for disease complications. Gut. 2002;51:316–322.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Moons LM, Kusters JG, van Delft JH, et al. A pro-inflammatory genotype predisposes to Barrett’s esophagus. Carcinogenesis. 2008;29:926–931.CrossRefPubMed Moons LM, Kusters JG, van Delft JH, et al. A pro-inflammatory genotype predisposes to Barrett’s esophagus. Carcinogenesis. 2008;29:926–931.CrossRefPubMed
27.
Zurück zum Zitat Izakovicova Holla L, Borilova Linhartova P, Hrdlickova B, et al. Haplotypes of the IL-1 gene cluster are associated with gastroesophageal reflux disease and Barrett’s esophagus. Human Immunol. 2013;74:1161–1169.CrossRef Izakovicova Holla L, Borilova Linhartova P, Hrdlickova B, et al. Haplotypes of the IL-1 gene cluster are associated with gastroesophageal reflux disease and Barrett’s esophagus. Human Immunol. 2013;74:1161–1169.CrossRef
28.
Zurück zum Zitat Gough MD, Ackroyd R, Majeed AW, Bird NC. Prediction of malignant potential in reflux disease: are cytokine polymorphisms important? Am J Gastroenterology. 2005;100:1012–1018.CrossRef Gough MD, Ackroyd R, Majeed AW, Bird NC. Prediction of malignant potential in reflux disease: are cytokine polymorphisms important? Am J Gastroenterology. 2005;100:1012–1018.CrossRef
29.
Zurück zum Zitat Babar M, Ryan AW, Anderson LA, et al. Genes of the interleukin-18 pathway are associated with susceptibility to Barrett’s esophagus and esophageal adenocarcinoma. Am J Gastroenterol. 2012;107:1331–1341.CrossRefPubMed Babar M, Ryan AW, Anderson LA, et al. Genes of the interleukin-18 pathway are associated with susceptibility to Barrett’s esophagus and esophageal adenocarcinoma. Am J Gastroenterol. 2012;107:1331–1341.CrossRefPubMed
30.
Zurück zum Zitat Gaj P, Mikula M, Wyrwicz LS, Regula J, Ostrowski J. Barrett’s esophagus associates with a variant of IL23R gene. Acta Biochim Pol. 2008;55:365–369.PubMed Gaj P, Mikula M, Wyrwicz LS, Regula J, Ostrowski J. Barrett’s esophagus associates with a variant of IL23R gene. Acta Biochim Pol. 2008;55:365–369.PubMed
31.
Zurück zum Zitat Casson AG, Zheng Z, Evans SC, et al. Cyclin D1 polymorphism (G870A) and risk for esophageal adenocarcinoma. Cancer. 2005;104:730–739.CrossRefPubMed Casson AG, Zheng Z, Evans SC, et al. Cyclin D1 polymorphism (G870A) and risk for esophageal adenocarcinoma. Cancer. 2005;104:730–739.CrossRefPubMed
32.
Zurück zum Zitat Su Z, Gay LJ, Strange A, et al. Common variants at the MHC locus and at chromosome 16q24.1 predispose to Barrett’s esophagus. Nat Genet. 2012;44:1131–1136.CrossRefPubMed Su Z, Gay LJ, Strange A, et al. Common variants at the MHC locus and at chromosome 16q24.1 predispose to Barrett’s esophagus. Nat Genet. 2012;44:1131–1136.CrossRefPubMed
33.
Zurück zum Zitat Palles C, Chegwidden L, Li X, et al. Polymorphisms near TBX5 and GDF7 are associated with increased risk for Barrett’s esophagus. Gastroenterology. 2015;148:367–378.CrossRefPubMedPubMedCentral Palles C, Chegwidden L, Li X, et al. Polymorphisms near TBX5 and GDF7 are associated with increased risk for Barrett’s esophagus. Gastroenterology. 2015;148:367–378.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Dura P, van Veen EM, Salomon J, et al. Barrett associated MHC and FOXF1 variants also increase esophageal carcinoma risk. Int J Cancer. 2013;133:1751–1755.CrossRefPubMed Dura P, van Veen EM, Salomon J, et al. Barrett associated MHC and FOXF1 variants also increase esophageal carcinoma risk. Int J Cancer. 2013;133:1751–1755.CrossRefPubMed
36.
Zurück zum Zitat Bird-Lieberman EL, Dunn JM, Coleman HG, et al. Population-based study reveals new risk-stratification biomarker panel for Barrett’s esophagus. Gastroenterology. 2012;143:927-e3–935-e3.CrossRef Bird-Lieberman EL, Dunn JM, Coleman HG, et al. Population-based study reveals new risk-stratification biomarker panel for Barrett’s esophagus. Gastroenterology. 2012;143:927-e3–935-e3.CrossRef
37.
Zurück zum Zitat Sikkema M, Kerkhof M, Steyerberg EW, et al. Aneuploidy and overexpression of Ki67 and p53 as markers for neoplastic progression in Barrett’s esophagus: a case–control study. Am J Gastroenterol. 2009;104:2673–2680.CrossRefPubMed Sikkema M, Kerkhof M, Steyerberg EW, et al. Aneuploidy and overexpression of Ki67 and p53 as markers for neoplastic progression in Barrett’s esophagus: a case–control study. Am J Gastroenterol. 2009;104:2673–2680.CrossRefPubMed
38.
Zurück zum Zitat Galipeau PC, Li X, Blount PL, et al. NSAIDs modulate CDKN2A, TP53, and DNA content risk for progression to esophageal adenocarcinoma. PLoS Med. 2007;4:e67.CrossRefPubMedPubMedCentral Galipeau PC, Li X, Blount PL, et al. NSAIDs modulate CDKN2A, TP53, and DNA content risk for progression to esophageal adenocarcinoma. PLoS Med. 2007;4:e67.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Maley CC, Galipeau PC, Li X, et al. The combination of genetic instability and clonal expansion predicts progression to esophageal adenocarcinoma. Cancer Res. 2004;64:7629–7633.CrossRefPubMed Maley CC, Galipeau PC, Li X, et al. The combination of genetic instability and clonal expansion predicts progression to esophageal adenocarcinoma. Cancer Res. 2004;64:7629–7633.CrossRefPubMed
40.
Zurück zum Zitat Yu JM, Yang LH, Guo Q, et al. Flow cytometric analysis DNA content in esophageal carcinoma. Correlation with histologic and clinical features. Cancer. 1989;64:80–82.CrossRefPubMed Yu JM, Yang LH, Guo Q, et al. Flow cytometric analysis DNA content in esophageal carcinoma. Correlation with histologic and clinical features. Cancer. 1989;64:80–82.CrossRefPubMed
41.
Zurück zum Zitat Watson A, Shepherd NA. The Definition of “Barrett’s” Columnar-Lined Oesophagus. British Society of Gastroenterology: Guidelines for the Diagnosis and Management of Barrett’s Columnar-Lined Oesophagus. 2005. Watson A, Shepherd NA. The Definition of “Barrett’s” Columnar-Lined Oesophagus. British Society of Gastroenterology: Guidelines for the Diagnosis and Management of Barrett’s Columnar-Lined Oesophagus. 2005.
42.
Zurück zum Zitat Buas MF, Levine DM, Makar KW, et al. Integrative post-genome-wide association analysis of CDKN2A and TP53 SNPs and risk of esophageal adenocarcinoma. Carcinogenesis. 2014;35:2740–2747.CrossRefPubMedPubMedCentral Buas MF, Levine DM, Makar KW, et al. Integrative post-genome-wide association analysis of CDKN2A and TP53 SNPs and risk of esophageal adenocarcinoma. Carcinogenesis. 2014;35:2740–2747.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Ren D, Zheng G, Bream S, Tevebaugh W, Shaheen NJ, Chen X. Single Nucleotide Polymorphisms of Caudal Type Homeobox 1 and 2 Are Associated with Barrett’s Esophagus. Dig Dis Sci. 2014;59:57–63.CrossRefPubMedPubMedCentral Ren D, Zheng G, Bream S, Tevebaugh W, Shaheen NJ, Chen X. Single Nucleotide Polymorphisms of Caudal Type Homeobox 1 and 2 Are Associated with Barrett’s Esophagus. Dig Dis Sci. 2014;59:57–63.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat van de Winkel A, Menke V, Capello A, et al. Expression, localization and polymorphisms of the nuclear receptor PXR in Barrett’s esophagus and esophageal adenocarcinoma. BMC Gastroenterol. 2011;11:108.CrossRefPubMedPubMedCentral van de Winkel A, Menke V, Capello A, et al. Expression, localization and polymorphisms of the nuclear receptor PXR in Barrett’s esophagus and esophageal adenocarcinoma. BMC Gastroenterol. 2011;11:108.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Murphy SJ, Hughes AE, Patterson CC, et al. A population-based association study of SNPs of GSTP1, MnSOD, GPX2 and Barrett’s esophagus and esophageal adenocarcinoma. Carcinogenesis. 2007;28:1323–1328.CrossRefPubMed Murphy SJ, Hughes AE, Patterson CC, et al. A population-based association study of SNPs of GSTP1, MnSOD, GPX2 and Barrett’s esophagus and esophageal adenocarcinoma. Carcinogenesis. 2007;28:1323–1328.CrossRefPubMed
46.
Zurück zum Zitat Casson AG, Zheng Z, Porter GA, Guernsey DL. Genetic polymorphisms of microsomal epoxide hydroxylase and glutathione S-transferases M1, T1 and P1, interactions with smoking, and risk for esophageal (Barrett) adenocarcinoma. Cancer Detect Prev. 2006;30:423–431.CrossRefPubMed Casson AG, Zheng Z, Porter GA, Guernsey DL. Genetic polymorphisms of microsomal epoxide hydroxylase and glutathione S-transferases M1, T1 and P1, interactions with smoking, and risk for esophageal (Barrett) adenocarcinoma. Cancer Detect Prev. 2006;30:423–431.CrossRefPubMed
47.
Zurück zum Zitat van Lieshout EM, Tiemessen DM, Witteman BJ, Jansen JB, Peters WH. Low glutathione and glutathione S-transferase levels in Barrett’s esophagus as compared to normal esophageal epithelium. Jpn J Cancer Res Gann. 1999;90:81–85.CrossRefPubMed van Lieshout EM, Tiemessen DM, Witteman BJ, Jansen JB, Peters WH. Low glutathione and glutathione S-transferase levels in Barrett’s esophagus as compared to normal esophageal epithelium. Jpn J Cancer Res Gann. 1999;90:81–85.CrossRefPubMed
48.
Zurück zum Zitat Casson AG, Zheng Z, Evans SC, Veugelers PJ, Porter GA, Guernsey DL. Polymorphisms in DNA repair genes in the molecular pathogenesis of esophageal (Barrett) adenocarcinoma. Carcinogenesis. 2005;26:1536–1541.CrossRefPubMed Casson AG, Zheng Z, Evans SC, Veugelers PJ, Porter GA, Guernsey DL. Polymorphisms in DNA repair genes in the molecular pathogenesis of esophageal (Barrett) adenocarcinoma. Carcinogenesis. 2005;26:1536–1541.CrossRefPubMed
49.
Zurück zum Zitat Ferguson HR, Wild CP, Anderson LA, et al. No association between hOGG1, XRCC1, and XPD polymorphisms and risk of reflux esophagitis, Barrett’s esophagus, or esophageal adenocarcinoma: results from the factors influencing the Barrett’s adenocarcinoma relationship case-control study. Cancer Epidemiol Biomark Prev. 2008;17:736–739.CrossRef Ferguson HR, Wild CP, Anderson LA, et al. No association between hOGG1, XRCC1, and XPD polymorphisms and risk of reflux esophagitis, Barrett’s esophagus, or esophageal adenocarcinoma: results from the factors influencing the Barrett’s adenocarcinoma relationship case-control study. Cancer Epidemiol Biomark Prev. 2008;17:736–739.CrossRef
50.
Zurück zum Zitat Menke V, van Zoest KP, Moons LM, et al. NcoI TNF-beta gene polymorphism and TNF expression are associated with an increased risk of developing Barrett’s esophagus and esophageal adenocarcinoma. Scand J Gastroenterol. 2012;47:378–386.CrossRefPubMed Menke V, van Zoest KP, Moons LM, et al. NcoI TNF-beta gene polymorphism and TNF expression are associated with an increased risk of developing Barrett’s esophagus and esophageal adenocarcinoma. Scand J Gastroenterol. 2012;47:378–386.CrossRefPubMed
51.
Zurück zum Zitat Menke V, Van Zoest KP, Moons LM, et al. Myo9B is associated with an increased risk of Barrett’s esophagus and esophageal adenocarcinoma. Scand J Gastroenterol. 2012;47:1422–1428.CrossRefPubMed Menke V, Van Zoest KP, Moons LM, et al. Myo9B is associated with an increased risk of Barrett’s esophagus and esophageal adenocarcinoma. Scand J Gastroenterol. 2012;47:1422–1428.CrossRefPubMed
52.
Zurück zum Zitat Bradbury PA, Zhai R, Hopkins J, et al. Matrix metalloproteinase 1, 3 and 12 polymorphisms and esophageal adenocarcinoma risk and prognosis. Carcinogenesis. 2009;30:793–798.CrossRefPubMedPubMedCentral Bradbury PA, Zhai R, Hopkins J, et al. Matrix metalloproteinase 1, 3 and 12 polymorphisms and esophageal adenocarcinoma risk and prognosis. Carcinogenesis. 2009;30:793–798.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat di Martino E, Hardie LJ, Wild CP, et al. The NAD(P)H:quinone oxidoreductase I C609T polymorphism modifies the risk of Barrett esophagus and esophageal adenocarcinoma. Genetics Med. 2007;9:341–347.CrossRef di Martino E, Hardie LJ, Wild CP, et al. The NAD(P)H:quinone oxidoreductase I C609T polymorphism modifies the risk of Barrett esophagus and esophageal adenocarcinoma. Genetics Med. 2007;9:341–347.CrossRef
54.
Zurück zum Zitat Rajendra S, Ackroyd R, Murad S, et al. Human leucocyte antigen determinants of susceptibility to Barrett’s oesophagus in Asians–a preliminary study. Aliment Pharmacol Ther. 2005;21:1377–1383.CrossRefPubMed Rajendra S, Ackroyd R, Murad S, et al. Human leucocyte antigen determinants of susceptibility to Barrett’s oesophagus in Asians–a preliminary study. Aliment Pharmacol Ther. 2005;21:1377–1383.CrossRefPubMed
55.
Zurück zum Zitat Kadioglu E, Sardas S, Ergun M, Unal S, Karakaya AE. The role of oxidative DNA damage, DNA repair, GSTM1, SOD2 and OGG1 polymorphisms in individual susceptibility to Barrett’s esophagus. Toxicol Ind Health. 2010;26:67–79.CrossRefPubMed Kadioglu E, Sardas S, Ergun M, Unal S, Karakaya AE. The role of oxidative DNA damage, DNA repair, GSTM1, SOD2 and OGG1 polymorphisms in individual susceptibility to Barrett’s esophagus. Toxicol Ind Health. 2010;26:67–79.CrossRefPubMed
56.
Zurück zum Zitat van Lieshout EM, Roelofs HM, Dekker S, et al. Polymorphic expression of the glutathione S-transferase P1 gene and its susceptibility to Barrett’s esophagus and esophageal carcinoma. Cancer Res. 1999;59:586–589.PubMed van Lieshout EM, Roelofs HM, Dekker S, et al. Polymorphic expression of the glutathione S-transferase P1 gene and its susceptibility to Barrett’s esophagus and esophageal carcinoma. Cancer Res. 1999;59:586–589.PubMed
57.
Zurück zum Zitat Chao DL, Sanchez CA, Galipeau PC, et al. Cell proliferation, cell cycle abnormalities, and cancer outcome in patients with Barrett’s esophagus: a long-term prospective study. Clin Cancer Res. 2008;14:6988–6995.CrossRefPubMedPubMedCentral Chao DL, Sanchez CA, Galipeau PC, et al. Cell proliferation, cell cycle abnormalities, and cancer outcome in patients with Barrett’s esophagus: a long-term prospective study. Clin Cancer Res. 2008;14:6988–6995.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Fang M, Lew E, Klein M, Sebo T, Su Y, Goyal R. DNA abnormalities as marker of risk for progression of Barrett’s esophagus to adenocarcinoma: image cytometric DNA analysis in formalin-fixed tissues. Am J Gastroenterol. 2004;99:1887–1894.CrossRefPubMed Fang M, Lew E, Klein M, Sebo T, Su Y, Goyal R. DNA abnormalities as marker of risk for progression of Barrett’s esophagus to adenocarcinoma: image cytometric DNA analysis in formalin-fixed tissues. Am J Gastroenterol. 2004;99:1887–1894.CrossRefPubMed
59.
Zurück zum Zitat Rabinovitch PS, Longton G, Blount PL, Levine DS, Reid BJ. Predictors of progression in Barrett’s esophagus III: baseline flow cytometric variables. Am J Gastroenterol. 2001;96:3071–3083.CrossRefPubMedPubMedCentral Rabinovitch PS, Longton G, Blount PL, Levine DS, Reid BJ. Predictors of progression in Barrett’s esophagus III: baseline flow cytometric variables. Am J Gastroenterol. 2001;96:3071–3083.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Teodori L, Gohde W, Persiani M, et al. DNA/protein flow cytometry as a predictive marker of malignancy in dysplasia-free Barrett’s esophagus: thirteen-year follow-up study on a cohort of patients. Cytometry. 1998;34:257–263.CrossRefPubMed Teodori L, Gohde W, Persiani M, et al. DNA/protein flow cytometry as a predictive marker of malignancy in dysplasia-free Barrett’s esophagus: thirteen-year follow-up study on a cohort of patients. Cytometry. 1998;34:257–263.CrossRefPubMed
61.
62.
Zurück zum Zitat Paulson TG, Maley CC, Li X, et al. Chromosomal instability and copy number alterations in Barrett’s esophagus and esophageal adenocarcinoma. Clin Cancer Res. 2009;15:3305–3314.CrossRefPubMedPubMedCentral Paulson TG, Maley CC, Li X, et al. Chromosomal instability and copy number alterations in Barrett’s esophagus and esophageal adenocarcinoma. Clin Cancer Res. 2009;15:3305–3314.CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Reid BJ, Prevo LJ, Galipeau PC, et al. Predictors of progression in Barrett’s esophagus II: baseline 17p (p53) loss of heterozygosity identifies a patient subset at increased risk for neoplastic progression. Am J Gastroenterol. 2001;96:2839–2848.CrossRefPubMedPubMedCentral Reid BJ, Prevo LJ, Galipeau PC, et al. Predictors of progression in Barrett’s esophagus II: baseline 17p (p53) loss of heterozygosity identifies a patient subset at increased risk for neoplastic progression. Am J Gastroenterol. 2001;96:2839–2848.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Dolan K, Walker SJ, Gosney J, Field JK, Sutton R. TP53 mutations in malignant and premalignant Barrett’s esophagus. Dis Esophagus. 2003;16:83–89.CrossRefPubMed Dolan K, Walker SJ, Gosney J, Field JK, Sutton R. TP53 mutations in malignant and premalignant Barrett’s esophagus. Dis Esophagus. 2003;16:83–89.CrossRefPubMed
65.
Zurück zum Zitat Dunn JM, Mackenzie GD, Oukrif D, et al. Image cytometry accurately detects DNA ploidy abnormalities and predicts late relapse to high-grade dysplasia and adenocarcinoma in Barrett’s oesophagus following photodynamic therapy. Br J Cancer. 2010;102:1608–1617.CrossRefPubMedPubMedCentral Dunn JM, Mackenzie GD, Oukrif D, et al. Image cytometry accurately detects DNA ploidy abnormalities and predicts late relapse to high-grade dysplasia and adenocarcinoma in Barrett’s oesophagus following photodynamic therapy. Br J Cancer. 2010;102:1608–1617.CrossRefPubMedPubMedCentral
Metadaten
Titel
Genetic Biomarkers of Barrett’s Esophagus Susceptibility and Progression to Dysplasia and Cancer: A Systematic Review and Meta-Analysis
verfasst von
John M. Findlay
Mark R. Middleton
Ian Tomlinson
Publikationsdatum
01.01.2016
Verlag
Springer US
Erschienen in
Digestive Diseases and Sciences / Ausgabe 1/2016
Print ISSN: 0163-2116
Elektronische ISSN: 1573-2568
DOI
https://doi.org/10.1007/s10620-015-3884-5

Weitere Artikel der Ausgabe 1/2016

Digestive Diseases and Sciences 1/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

RAS-Blocker bei Hyperkaliämie möglichst nicht sofort absetzen

14.05.2024 Hyperkaliämie Nachrichten

Bei ausgeprägter Nierenfunktionsstörung steigen unter der Einnahme von Renin-Angiotensin-System(RAS)-Hemmstoffen nicht selten die Serumkaliumspiegel. Was in diesem Fall zu tun ist, erklärte Prof. Jürgen Floege beim diesjährigen Allgemeinmedizin-Update-Seminar.

Gestationsdiabetes: In der zweiten Schwangerschaft folgenreicher als in der ersten

13.05.2024 Gestationsdiabetes Nachrichten

Das Risiko, nach einem Gestationsdiabetes einen Typ-2-Diabetes zu entwickeln, hängt nicht nur von der Zahl, sondern auch von der Reihenfolge der betroffenen Schwangerschaften ab.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Battle of Experts: Sport vs. Spritze bei Adipositas und Typ-2-Diabetes

11.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Im Battle of Experts traten zwei Experten auf dem Diabeteskongress gegeneinander an: Die eine vertrat die Auffassung „Sport statt Spritze“ bei Adipositas und Typ-2-Diabetes, der andere forderte „Spritze statt Sport!“ Am Ende waren sie sich aber einig: Die Kombination aus beidem erzielt die besten Ergebnisse.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.