Skip to main content
Annals of Oncology logoLink to Annals of Oncology
. 2014 Sep 11;26(4):624–644. doi: 10.1093/annonc/mdu449

A systematic review and meta-analysis of somatic and germline DNA sequence biomarkers of esophageal cancer survival, therapy response and stage

J M Findlay 1,2, M R Middleton 3, I Tomlinson 1,3,*
PMCID: PMC4374384  PMID: 25214541

Recent advances in next generation sequencing reinforce the potential for DNA sequence markers to guide esophageal cancer management. We report the first systematic review and meta-analysis, identifying 94 markers of outcome and 41 of stage. Overall, evidence was poor. Meta-analyses demonstrated outcome associations for 6 tumor and 9 germline variants: priorities for prospective evaluation.

Keywords: biomarkers, genomic, cancer, esophageal, prognosis, cancer staging

Abstract

Introduction

There is an urgent need for biomarkers to help predict prognosis and guide management of esophageal cancer. This review identifies, evaluates and meta-analyses the evidence for reported somatic and germline DNA sequence biomarkers of outcome and stage.

Methods

A systematic review was carried out of the PubMed, EMBASE and Cochrane databases (20 August 2014), in conjunction with the ASCO Level of Evidence scale for biomarker research. Meta-analyses were carried out for all reported markers associated with outcome measures by more than one study.

Results

Four thousand and four articles were identified, 762 retrieved and 182 studies included. There were 65 reported markers of survival or recurrence 12 (18.5%) were excluded due to multiple comparisons. Following meta-analysis, significant associations were seen for six tumor variants (mutant TP53 and PIK3CA, copy number gain of ERBB2/HER2, CCND1 and FGF3, and chromosomal instability/ploidy) and seven germline polymorphisms: ERCC1 rs3212986, ERCC2 rs1799793, TP53 rs1042522, MDM2 rs2279744, TYMS rs34743033, ABCB1 rs1045642 and MTHFR rs1801133. Twelve germline markers of treatment complications were reported; 10 were excluded. Two tumor and 15 germline markers (11 excluded) of chemo (radio)therapy response were reported. Following meta-analysis, associations were demonstrated for mutant TP53, ERCC1 rs11615 and XRCC1 rs25487. There were 41 tumor/germline reported markers of stage; 27 (65.9%) were excluded.

Conclusions

Numerous DNA markers of outcome and stage have been reported, yet few are backed by high-quality evidence. Despite this, a small number of variants appear reliable. These merit evaluation in prospective trials, within the context of high-throughput sequencing and gene expression.

introduction

Esophageal and gastroesophageal junctional (GEJ) carcinoma account for 3.9% of cancer diagnoses yet 5.9% of cancer deaths [1]. Worldwide, squamous cell carcinoma (SCC) predominates but, in Western countries, incidence of adenocarcinoma is increasing rapidly [2, 3]. Treatment with curative intent involves either resection with or without neoadjuvant therapy, or definitive chemoradiotherapy with or without salvage resection. More than 5000 patients undergo esophagectomy in the United States and the UK every year, with 85% receiving neoadjuvant therapy [4, 5]. However, the majority experience complications, operative mortality remains relatively high and quality of life may be significantly impaired [68]. Neoadjuvant, adjuvant and definitive chemo- and/or radiotherapy also carry risk [9], and while the absolute survival benefit of neoadjuvant therapy ranges from 7% to 13% at 2 years [9], 50%–60% of tumors are resistant [10].

Prognosis overall remains bleak; even following ostensibly curative treatment 5-year survival is just 35%–45% [1113]. This highlights limitations in our biological understanding, and our urgent need for biomarkers to predict prognosis, recurrence and sensitivity to therapy, and ultimately better personalize care. Most clinical experience with esophageal biomarkers to date has largely involved protein expression with or without sequence changes; while such markers are used to select patients for early phase trials, the sole tumor marker in routine use is ERBB2/HER2 status [14, 15]. However, rapid advances in high-throughput next-generation sequencing (NGS) have highlighted the potential role of somatic DNA sequence markers. These may function as independent markers, serve to refine or explore existing expression markers, or constitute novel therapeutic targets [1618]. Similarly, advances in custom and genome-wide single nucleotide polymorphism (SNP) arrays have emphasized the role of germline variants in modulating cancer and treatment outcome [19, 20].

We therefore undertook the first systematic review of DNA sequence biomarkers of esophageal cancer, to systematically identify and evaluate all candidate somatic and germline DNA sequence markers of outcome (survival, recurrence, therapy response and treatment complications) and stage. We then performed meta-analysis for all markers with a nominally statistical association in at least one study.

methods

inclusion criteria

Studies eligible were those testing association between a DNA sequence marker (germline or somatic) and outcome (clinical, radiological or pathological) or stage of esophageal/GEJ cancer. Markers included germline SNPs, tumor mutations, copy number variants (CNVs), loss of heterozygosity (LOH), microsatellite instability (MSI) and chromosomal instability (CIN; alterations in ploidy). Clinical outcomes comprised survival (any measure), recurrence, disease progression and treatment complications. Radiological outcomes comprised response to therapy. Histopathological outcomes comprised tumor response and incomplete resection. Stage comprised radiological TNM staging and pathological tumor grading [21].

exclusion criteria

Studies using cell lines or expression data were excluded unless discrete tumor or DNA-specific data could be extracted. Non-English articles were excluded.

literature search

A search was performed on 20 August 2014 of the PubMed, EMBASE and Cochrane databases, in accordance with MOOSE (Meta-analysis Of Observational Studies in Epidemiology) and PRISMA guidelines [22]. The following term was used: (esophageal OR esophagus OR gastroesophageal) AND (cancer OR carcinoma or adenocarcinoma OR SCC) 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) AND (outcome OR prognosis OR survival OR response OR stage OR surgery OR chemotherapy OR radiotherapy OR marker OR biomarker OR complication). The references cited by retrieved articles were also assessed for relevant articles.

study data

Data extracted were: methodology; the variant(s) and gene(s) assessed; outcome measures and population. Extraction was carried out independently by two authors (JMF and IT). Gene names were standardized (HUGO Gene Nomenclature Committee) [23]. Variants were mapped to reference SNP (rs) identification numbers (US National Library of Medicine dbSNP database; http://www.ncbi.nlm.nih.gov/snp) when not provided by searching referenced methodology, in vitro polymerase chain reaction (http://genome.ucsc.edu) with specialized SNP flank BLAST® (Basic Local Alignment Search Tool; http://blast.ncbi.nlm.nih.gov), or New England Biocutter v2.0 (NEBcutter; http://tools.neb.com/NEBcutter2). Gene function was classified using the US National Library of Medicine Gene Database (http://www.ncbi.nlm.nih.gov/gene). For all reported associations, it was determined whether statistical significance persisted following correction for multiple comparisons (Bonferroni or false discovery rate correction), or multivariate analysis of all variables including genotypes. Were none made, post hoc Bonferroni correction was carried out [24]. For genome-wide association studies, significance was assumed at P < 5 × 10−8. For reported markers assessed by a single study, for which P was <0.05 but >corrected α, effect metrics were calculated but the marker excluded and presented in supplementary Tables S1 and S2, available at Annals of Oncology online. Those assessed by more than one study underwent meta-analysis irrespective.

evidence quality

Quality was appraised using the revised American Society of Clinical Oncology Level of Evidence (LOE) scale for biomarker research [25].

meta-analysis

Meta-analysis was carried out for all markers with a statistically significant association (uncorrected P < 0.05) reported by at least one study. For SNPs, analysis was carried out using the major common allele as reference, using genotype permutations shared by all studies. In the case of A/T and C/G substitutions, the minor variant was confirmed from study allele frequencies. Where possible, separate analyses were carried out for major methodological differences such as adjusted/unadjusted hazard ratios (HRs), genotyping methods, treatment, cell type and ethnicity (as determined by the International HapMap Consortium) [26]. Natural logarithms of HR, odds ratios (ORs) and standard errors (SEs) were extracted. In studies not presenting these, these were estimated using the methods of Parmar, or extracted from magnified Kaplan–Meier survival curves: HR and SE were estimated at constant time points; censoring was assumed to be constant and starting from the minimal follow-up period, with censored patients allocated to the appropriate time interval [27]. In six meta-analyzed studies (all nonsignificant results) [2833], it was not possible to extrapolate statistics for all variants; an lnHR of 0 (a HR of 1) and SE of the most closely matched study (regarding cell type, size and methodology) were used to minimize selection bias. When not presented, ORs were calculated from available data. Meta-analysis was carried out using RevMan v5.2 (Copenhagen: the Nordic Cochrane Centre, The Cochrane Collaboration).

study heterogeneity and bias

Heterogeneity was quantified using I2 and χ2 estimates; for moderate heterogeneity (I2 ≥ 50%) random rather than fixed-effects models were used. Heterogeneity and bias were also assessed by funnel plot asymmetry; [34] visually for all analyses, and statistically for analyses involving at least 10 studies [34, 35] using Begg's and Egger's tests. Statistical significance was assumed at P < 0.05. Following consideration of alternative causes, probable publication bias was corrected using the ‘trim and fill’ method [36]. All other analysis was carried out using R (v3.0.2) [37]. Sensitivity analyses were carried out for all analyses including five studies or more, whereby studies were omitted one by one.

results

study characteristics

Four thousand and four articles were identified, 762 retrieved for evaluation, 580 excluded (Figure 1) and 184 included (supplementary Tables S3–S25, available at Annals of Oncology online), published between 1989 and 2014. Seventy-three assessed markers of clinical outcome, 80 clinical outcome and stage and 29 stage alone. Survival measures were overall survival (OS; n = 133), disease-free survival (DFS; n = 20), recurrence (n = 19), progression-free survival (PFS; n = 4) and disease-specific survival (DSS; n = 4). Twenty nine studies assessed response to therapy (chemo ± radiotherapy, or biological). Eleven assessed treatment complications. Treatment intent was curative (n = 156), palliative (n = 5), mixed (n = 21) and unspecified in 1. Curative modalities were resection alone (n = 111), resection ± neoadjuvant (n = 33) or adjuvant (n = 2) therapy, or definitive chemoradiotherapy (n = 9). All chemotherapy regimens involved platinum agents with or without 5-fluorouracil, except three (bleomycin, gefitinib, irinotecan). One hundred and seventy-six studies were candidate based, and 6 genome-wide (1 SNP, 5 CNV). One hundred and seventeen studies assessed tumor variants, 64 germline and 1 both. Cell types assessed were SCC (n = 117), adenocarcinoma (AC) (n = 40), both (n = 3) and unspecified (n = 22).

Figure 1.

Figure 1.

PRISMA flow diagram.

methodological quality

LOE was B for five studies (2.75%), C for 104 (57.1%) and D for 73 (40.1%). Median number of subjects was 90 (range 10–2932), although 48 studies included fewer than 50. Forty-six (25.3%) studies were prospective; 135 (74.7%) were retrospective; 1 (0.55%) had both components. Multivariate adjustment of effect sizes was carried out by 57 studies (31.3%).

molecular quality

Just 37 (56.3%) of 65 studies assessing germline variants assessed Hardy–Weinberg equilibrium; 59 (90.8%) reported genotyping success rate (or provided data allowing its calculation).

markers of survival and recurrence

There were 65 reported markers of survival or recurrence: 24 tumor (Table 1; 3 mutations, 16 CNV, 2 LOH regions, 1 telomere length ratio, CIN, heterogeneous ploidy) and 40 germline polymorphisms (Table 2).

Table 1.

Reported tumor markers (mutations, copy number variants, and chromosomal instability) associated with survival and recurrence following treatment of esophageal cancer

LOE Variant Gene / function Association – minor variant Association – wild type No association Cell type Population LOE Meta-HR [effect variant] Chi I2 N P
Mutations
III Exon mutant TP53
Apoptotic / DNA repair regulator
OS and DFS Casson 2003 [38]
OSA – Schneider 2000 [39]
OS and DFS–Madani 2010 [40]


OSA – Yamasaki 2010 [41]
OSA – Kunisaki 2006
OS – Kobayashi 1999 [42]

OS – Uchino 1996 [29]









OS and DFS – Ribeiro 1998 [43]
OS and DSS – Kandioler 2014 [44]



OS – Puhringer 2006 [45]
OS – Soontrapornchai 1999 [46]



OS – Makino 2010 [47]

OS – Shimada 1997 [48]
OS – Egashira 2011 [49]
OS – Ito 2001 [50]
OS – Lam 1997 [51]
OS – Shibagaki 1995 [52]
OS: Goan 2005 [53]
OS: Cao 2004 [54]
OSA – Gibson 2003 [55]
OSA – Coggi 1997 [56]
AC
AC
AC
AC
AC
SCC
SCC
SCC
SCC
SCC
SCC
SCC
SCC
SCC
SCC
SCC
SCC
US
US
US
US
Res
Res
Res
Res + /−NAC(RT) (CF)
Res
Mixed
Res + NAC
Res+ NAC
Res + NACRT (CF)
Res
Res
Res
DCRT
Res
Res
Res
Res
Res + NACRT
Res
Res + NACRT (CF + IFN)
Res + NAC (CF)
C
C
C
B
D
D
C
D
D
D
C
C
C
D
C
D
C
C
D
D
C
OS(A)PB: 1.27 (1.01–1.59)
OSA PB: 1.19 (0.63–2.27)
AC(A): 1.99 (1.44–2.81)
SCC(APB: 1.47 (1.24–1.73)
US(A)PB: 0.77 (0.41–1.47)

DFS(A): 2.67 (1.38–5.15)
Res OS(A)PB: 1.35 (1.04–1.76)
NAC/RA(PB): 1.23 (0.81–1.87)

SSCP analysis
OS(A)PB: 1.56 (1.33–1.82)
Direct sequencing only
OS(A)PB: 0.96 (0.62–1.46)
SCCP band only
68.1
34.5
2.68
22.7
21.3

7.44
27.0
32.5


27.8

28.3
62
80
0
43
72

73
56
66


44

65
21
6
5
11
5

3
13
7


13

8
0.04
0.590
< 0.001
< 0.001
0.440

0.003
0.030
0.320


< 0.001

0.830
IV Exon 9/20 mutation PIK3CA
Cell signalling kinase
OSA and DFSA – Shigaki 2013 [58] Rec – Shigaki 2013

OSA and RecEx – Wang 2014
OSEx and DFSEx – Hou 2014
SCC

SCC
SCC
Res + /−NACRT (CF + /−tax)

Res + /−NACRT
Res
D

D
C
OS(A): 0.63 (0.26–1.56)

DFS(A): 0.42 (0.21–0.85)
RecEx: 0.64 (0.23–1.75)
6.13

0.26
2.81
67

0
64
3

2
2
0.320

0.020
0.390
IV Exon mutation NRF2/BIRC2
Transcription factor
OS and Rec – Shibata 2011 SCC Resection + NACRT (F) (Japan) D
OSEx: 3.54 (1.60–7.88)
Rec—NP
NA NA 1 0.005
0.046
Copy number variants
II Gain EGFR
Epidermal growth factor receptor
OS – Marx 2010 [59]
OS – Kitagawa 1996 [60]




OS – Lennerz 2011 [61]







OSA – Luber 2011 [62]


OS – Miller 2003 [28]
OS – Janmaat 2006 [63]
OS Rec – Chikuba 1995 [30]
OS –Itakura 1994 [31]


OS – Sunpaweravong 2005 [64]
AC
SCC
AC
SCC
SCC
SCC
US
AC
SCC
Res
Res
Res
Palliative gefitinib
Res + ACRT (C)
Res
Mixed
PC (OLF + cetuximab)
Res
D
D
C
B
D
D
D
B
D
Gain assessed by FISH/CISH
OS: 2.43 (0.75–7.84)

Gain assessed by slot/Southern blot
OS: 1.63 (0.63–4.22)

Excluding Miller 2003 (qPCR)

23.9


18.1

92


89

3


3

0.140


0.320
III Gain ERBB2/HER2
Epidermal growth factor receptor

OS – Prins 2013 [65]

DFSA – Rauser 2007 [66]
OSA – Brien 2000 [67]



OS – Zhan 2012 [68]
OS – Sato-Kuwabara 2009 [69]
OSA – Mimura 2005 [70]


DSSA, OSA – Yoon 2012 [71]
OSA – Rauser 2007 [66]

OS – Thompson 2011 [72]
OS – Miller 2003 [28]
OS – König 2013 [73]



OS – Sunpawerayong 2005 [64]
OS – Suzuki 1997 [74]
OS and DSS Ikeda 1996 [75]
OS – Lennerz 2011 [61]

AC
AC
AC
AC
AC
AC
Both
SCC
SCC
SCC
SCC
SCC
SCC
US

Res
Res
Res
Res
Res
Res


Res
Res
Res
Res
Ress
Mixed

D
C
D
D
D
C
D
D
D
D
C
D
D
D

OS(A): 1.63 (1.20–2.21)
OSA: 2.31 (1.64–3.24)
OS(A) (AC): 1.59 (0.99–2.56)
OS(A) (SCC): 1.92 (1.12–3.29)
DFSA: 2.1 (1.06–4.26)

Gain assessed via
FISH/SISH/IHC; Miller 2003,
Ideka 1996, Suzuki 1997
excluded

31.2
0.72
15.4
27.7
NA

62
0
68
82
NA

11
3
6
5
1

0.002
< 0.001
0.060
0.020
0.033
III Gain ERBB2/HER2
Epidermal growth factor receptor
DSSA, OSA – Yoon 2012 [71]
(heterogeneous amplification)
AC Res C OS: 2.02 (1.09–3.74)
DSS: 2.04 (1.09–3.79)
NA
NA
NA
NA
1
1
0.026
0.025
III Gain CCND1
Cell cycle kinase
OSA: Wang 2012b [76]
OSA – Miller 2003 [28]
OSA – Takeshita 2010 [77]


OS – Shimada 1997 [48]
OS – Shinozaki 1996 [78]
RecA – Komatsu 2014 [79] (ctDNA)



OS–Sunpawerayong 2005 [64]
OS – Gramlich 1994 [80]
SCC
AC
SCC
SCC
SCC
SCC
SCC
SCC
Res
Res
Res
Res
Res
Res
Res
Res
C
C
C
D
C
D
D
C
Gain assessed by qPCR only
OSA: 2.09 (1.27–3.42)
Gain assessed by FISH/IHC
OSA: 1.54 (0.93–2.57)
Gain assessed by slot blot
OS: 4.29 (2.47–7.45)

3.17

3.94

NA

5

75

NA

4

2

1

0.004

0.100

< 0.001
III Gain 1p36.32 OSA – Carneiro 2008 [81] SCC Res C OSA: HR 19.6 (2.5–153.9) NA NA 1 0.005
III Gain 19p13.3 OSA – Carneiro 2008 [81] SCC Res C OSA: HR 7.0 (1.5–31.9) NA NA 1 0.011
III Gain MDM2
Ubiquitin ligase
OS – Shibagaki 1995 [52] SCC Res C OS: HREx 3.82 (1.81–8.07) NA NA 1 5.3x10−3
IV Gain FGF3/INT2
Fibroblast growth factor

OS – Ikeda 1996 [75]
OS – Mori 1992 [82]

OS – Shimada 1997 [48]
OS – Suzuki 1997 [74]
AC
SCC
SCC
SCC
Res
Res
Res
Res
D
D
D
D
OS: PBHR 1.83 (1.18–2.83)
PB – Corrected for Ikeda 1996
5.65 29 4 0.006
IV Gain FGF4/HST1
Fibroblast growth factor
OS – Chikuba 1995 [30] Rec – Chikuba 1995 [30] SCC
SCC
Res + ACRT (C)
Res
D
D
Median survival different but:
OS: HREx 1.4 (0.86–2.30)
Rec: HREx 1.09 (0.659–1.80)
NA NA 1 >0.05
IV Gain TERC
Telomerase
OS – Wang 2013 [83] SCC Res D OS: HREx 7.87 (3.32–18.7) NA NA 1 0.010
IV Gain MET
Growth factor
OS – Lennerz 2011 [61] US Mixed D OS: HREx 3.72 (2.56–5.39) NA NA 1 < 0.001
IV Gain CPT1A
Mitochondrial oxidation
OSA – Shi 2011 [84] SCC Res D OSA: 4.39 (1.34–14.14) NA NA 1 0.015
Telomere length
III Telomere length ratio (>1.17) OSA – Gertler 2008 [85] AC Res C OS: HRA 3.40 (1.3–8.9) NA NA 1 < 0.02
LOH
III LOH at one of 2p, 3p, 17p OS – Ikeguchi 1999 [86] SCC Res C 2 loci: OS (3yr) 48% versus 75%
HREx: 1.81 (0.53–6.25)
NA NA 1 0.048
> 0.05
IV LOH 1q21-23 OS – Maru 2009 [87] AC Res D HREx: 3.90 (1.13–13.5) NA NA 1 0.030
Chromosomal instability
III Aneuploid / polyploidy OS – Doki 1993 [88]
Rec – Tsutsui 1992 [89]
Rec – Kaketani 1989 [90]
OS – Ohno 1989 [91]
OS – Kuwano 1995 [92]





OSA – Wang 1999 [32]
OS Edwards 1989 [93]
SCC
SCC
SCC
SCC
SCC
SCC
SCC
Res
Res
Res
Res + NACRT
Res
Res
Res
D
D
D
C
C
D
D
PBOS(A)*: 1.63 (1.25–2.11)
Rec: 5.41 (0.87–33.8)
2.99
2.11
25
53
4
2
2x10−4
0.070
IV Ploidy heterogeneity (Homogeneity)
OS – Deguchi 1993 [94]
DFS- Deguchi 1993 [94] SCC Res D OS: 0.10 (0.03–0.36)
DFS: 0.34 (0.08–1.42)
NA
NA
NA 1 < 0.05

OS = overall survival; DFS = disease-free survival; rec = recurrence; A = adjusted; (A) = including adjusted; PB = adjusted for publication bias; Ex = extrapolated; NP = not presented; AC = adenocarcinoma; SCC = squamous cell carcinoma; US = unspecified; NAC = neoadjuvant chemotherapy; NACRT = neoadjuvant chemoradiotherapy; DCRT = definitive chemoradiotherapy; PC = palliative chemotherapy; Res = resection; C = cisplatin; F = 5FU; CFL = cisplatin-5FU-leucovorin; OFL = cisplatin-5FU-leucovorin;; tax = taxane; SSCP = single strand conformation polymorphism; ctDNA = circulating tumor DNA; NA = not applicable

Table 2.

Reported germline markers (polymorphisms) associated with survival and recurrence following treatment of esophageal cancer

LOE SNP
(major/minor allele)
Gene /
function
Association–
minor variant
Association–
wild type
Non-significant Cell type Population Meta-HR [variant allele / genotype] versus wild-type' [Ethnicity] Chi I2 N P
II rs3212986
(C/A)
ERCC1
DNA NER
OSA–Bradbury 2009b [95]
DFSA–Wang 2011 [96]
OSA–Rumiato 2013 [97]




OS–Warnecke 2009 [33]
US
SCC
US
US
Res + NAC (CF)
PC (CF)
Res +/− NAC (CF)
Res + NACRT (CF)
B

C
C
C
OS(A/Ex): 0.63 (0.42–0.93)
[TT/CT, cisplatin, Caucasian]


DFS(A): 1.98 (1.19–3.03)
[AA/CA + cis,,Chinese]
4.12



NA
51



NA
3



1
0.02



0.001
II rs1799793
(G/A)
ERCC2
DNA NER
OSA–Bradbury 2009b [95]

OS and Rec – Ott 2011 [98]

OS–Rumiato 2013 [97]
US
AC

US
Res + NAC (CF)
Res + NAC (C/OF +/−tax)
Res +/−NAC (CF)
B
B

C
OS(A/Ex): 0.71 (0.54–0.94)
[GA/AA; cisplatin; Caucasian]

DFS: 0.33 (0.20–0.07) [AA]
3.88


NA
48


NA
3


1
0.020


0.002
II rs13181
(T/G)
ERCC2
DNA NER
OSA and DFSA–Bradbury 2009b [95]
OS and Rec - Ott 2011 [98]

OS–Rumiato 2013 [97]
OSA, RecA–Wu 2006
US
AC

US
US
Res + NAC (CF)
Res + NAC (C/OF +/−tax)
Res +/−NAC (CF)
Res + NACRT (CF +/−tax)
B
B

C
D
OS(A/Ex): 0.82 (0.65–1.05)
[TG/GG, Caucasian]

DFSA: 0.32 (0.20–0.60)
RecA: 0.94 (0.30–2.81)
5.14


NA
0.03
42


NA
0
4


1
2
0.110


0.002
0.91
III rs11614913 MIR196A2
Micro RNA
OSA–Wu 2014 [99]

OSA–Yang 2014b [100]
SCC

SCC
PC

Mixed
C
C
OSA: 1.26 (0.72–2.21)
[TT; Chinese/Taiwanese]
PFSA: 1.01 (0.54–1.88)
2.17

NA
54

NA
2

1
0.420

0.972
III CA-SSR-1
(DNR)
EGFR
Epidermal growth factor receptor
OS and Rec–Vashist 2014 [101] Both Res C OS (AC): 1.70 (1.20–2.80)
[LL; Caucasian]
Rec (AC): 2.70 (1.70–4.30)
OS (SCC): 3.50 (2.10–6.00)
Rec (SCC): 2.50 (1.30–4.80)
NA NA 1 0.010

< 0.001
< 0.001
0.005
III rs1800796
(C/G)
IL6
Interleukin
OS–Motoyama 2012b [102] DSS–Motoyama 2012b [102] SCC Res +/− AC C OS: HRA 3.40 (CI NP)
[GG/GC; Japanese]
OS: HRAEx 3.49 (1.28–9.45)
NA NA 1 5.9x10−3

< 0.05
III rs238406
(G/T)
ERCC2
NER repair
OSA and DFSA Lee 2011 [103] SCC Res + NACRT (CF/C + tax) C OSA: 0.61 (0.40–0.93)
[CC; Taiwanese]
DFSA: 0.57 (0.38–0.85)
NA NA 1
0.02

0.007
III rs1800975
(G/A)
XPA
DNA NER repair
OSA–Yang 2013 [104] DFS–Yang 2013 [104]
SCC
Mixed
C
OSA: 1.36 (1.06–1.76) [AG;
[Taiwanese]
DFS: 1.20 (0.95–1.51) [AG]
NA
NA
1
0.014

0.126
III rs34743033
(STR 2/3/4)
TYMS
DNA repair/ replication
OSA–Kaneko 2011 [105]
OS–Okuno 2007 [106]
OS–Sarbia 2006 [107]

OS–Rumiato 2013
SCC
SCC
SCC

US
DCRT (CF)
Res + NACRT (CF)
Res + NACRT (CF + E)

Res + NAC (CF)
C
C
C

C
OSA: 1.54 (1.00–2.38)
[≥2/3; Caucasian/Japanese]
OS(A): 2.47(1.20, 5.06) [Japanese]

OSA: 1.18 (0.69, 2.03) [Caucasian]
5.78

0.45

0.18
65

0

0
3

2

2
0.61

0.010

0.550
III rs2279744
(T/G)
MDM2
Ubiquitin ligase



OSA–Renouf 2013 [108]
DFSA Boonstra 2011 [109]
OSA and DFSA Cescon 2009 [110]
Both
Both

AC
Res
Mixed
D
C
(SCC) DFSA: 2.78 (0.24–31.9) [GG; Caucasian]
(AC) DFSA: 0.92 (0.65–1.29) [GG]

(AC) OSA: 2.01 (1.38–2.95)
(SCC) OSA: 7.89 (2.40–26.0)
9.36


0.04
0.48
NA
89


0
0
NA
2


2
2
1
0.410


0.620
< 0.001
< 0.001
III rs2273535
(A/T)
AURKA
Cell cycle kinase
OS and DFSA–Pan 2012 [111]

DFSA–Boonstra 2011 [109]
US

Both
Res + NACRT (CF + tax)

Res
C

D
OSEx: 0.30 (0.10–0.92) [TT; Caucasian]

DFSA: 0.55 (0.17–1.74)
NA

2.92
NA

66
1

2
< 0.05

0.310
III rs2010963
(G/C)
VEGFA
Epithelial mitogen
OS–Tamura 2012 [112]
OS - Yang 2014 [113]
SCC
SCC
DCRT (CF)
Res +/−NACRT (CF)
C OS HR(Ex): 0.68 (0.50–0.92) [CC] 0.16 0 2 0.01
III rs3025039
(C/T)
VEGFA Epithelial mitogen DFSA–Lorenzen 2011 [114]

OSA–Bradbury 2009 [115]


OS–Tamura 2012 [112]
AC
AC
SCC
Res + NAC (CF)
Mixed
DCRT (CF)
C
C
C
OS(A): 0.75 (0.54–1.03) [CT; Caucasian/Japanese]
DFSA: 1.8 (1.04–3.09) [CT/TT; Caucasian]
0.79

NA
0

NA
2

1
0.080

0.040
III rs1042522
(C/G)
TP53
Apoptotic / DNA repair regulator
OSA and PFSA–Renouf 2013 [108]
OSA, DFSA–Cescon 2009 [110]



OSA, RecA–Wu 2006
AC

AC
US
Mixed

Mixed
Res + NACRT (CF +/−tax)
C

C
D
OSA: 1,84 (1.34–2.53) [GG; Caucasian]

DFSA: 2.03 (1.29–3.18) [GG]
RecA: 1.29 (0.24–7.14) [GG]
0.44

NA
NA
0

NA
NA
3

1
1
< 0.001

0.002
> 0.05
III rs2069762
(A/C)
IL2
Cytokine
DSSA–Motoyama 2011 [116] SCC Res C DSS: 3.54 1(1.69–7.39) [C; Japanese]
DSSA: 3.36 (NA)
NA NA 1 0.0231

0.0136
III rs1800471
(C/G)
TGFB1
Growth factor regulator
OS–Tang 2013 [117] SCC Mixed C OS: 3.51 (2.18–5.67) [CG/GG; Chinese] NA NA 1 < 0.001
III rs1050631
(G/A)
SLC39A6
Zinc transporter
OSA–Wu 2013 [118] SCC Mixed C OSA: 1.3 (1.19–1.43) [AA; Chinese] NA NA 1 3.77x10−8
III rs41458645
(C/T)
Mitochondrial D loop OSA–Zhang 2010 [119] SCC Mixed C OSA: 3.00 (1.03–8.76) [CT; Chinese] NA NA 1 0.044
III rs139001869
(A/G)
Mitochondrial D loop OSA–Zhang 2010 [119] SCC Mixed C OSA: 3.48 (1.07–11.36) [AG; Chinese] NA NA 1 0.039
III rs3769818
(G/A)
CASP8
Caspase
OSA –Umar 2011 [120] SCC Mixed C OSA: 3.36 (1.07–10.61) [AA; Indian] NA NA 1 0.039
III rs1695
(A/G)
GSTP1
Detoxification enxyme
OSA–Lee 2005 [121]

OS–Okuno 2007 [106]
OS–Warnecke 2009 [33]
OSA and RecA – Wu 2006 [122]
OSA – Rumiato 2013
SCC

SCC
US
US

US
Res + NAC
(C + F/tax)
Res + NACRT (CF)
Res + NACRT (CF)
Res + NACRT (CF +/−tax)
Res + NAC (CF)
C

C
C
D

C
OSA: 1.29 (1.03–1.61) [TG/GG; Caucasian/Taiwanese/Japanese]
OSA 1.15 (0.78–1.70)
[TG/GG; Caucasian]
RecA: 0.50 (0.16–1.58) [Caucasian]
2.36


1.84


NA
0


0


NA
5


3


1
0.030


0.490


> 0.05
III rs72214039
(CA ins)
EGFR
Epidermal growth factor receptor
OSA–Lee 2011b [123] SCC Res NACRT (CF/C + tax) C OSA: 1.88 (1.02–3.49) [Short/Short; Taiwanese] NA NA 1 0.045
III rs7121
(T/C)
GNAS
G protein subunit
OSA – Alakus 2014 [124]
OSA and DFSA–Vashist 2011 [125]



OS – Alakus 2009 [126]
US
US

US
Res
Res

Res + NACRT (CF)
D
C

B
OSA: 0.73 (0.46–1.16) [CC; Caucasian]
DFSA: 0.55 (0.34–0.89
9.08


NA
67


NA
3


1
0.180


2.50x10−3
III rs111509018
(STR)
ECRG2/SPINK7
Serpin-inhibitor
OSA and DFSA Kaifi 2007 [127] US Res C OSA: 2.56 (1.53–4.29) [TCA4/TCA4; Caucasian]
DFSA: 2.30 (1.37–3.87)
NA
NA 1

1
< 0.001

< 0.001
III rs9344
(G/A)
CCND1
Cell cycle kinase
OSA–Izzo 2007 [57] AC Res D OSA: 3.48 (1.94–6.23) [AA] NA NA 1 < 0.001
IV rs1801133
(G/A)
MTHFR
Folate metabolism
RecA–Wu 2006 [122] OSA–Wu 2006 [122]

OSA and RecA Ott 2011 [98]

OS – Lu 2011 [128]
OS–Umar 2010 [129]
OS–Sarbia 2006 [107]

OS–Warnecke 2009 [33]
US

AC

SCC
SCC
SCC

SCC
Res + NACRT (CF +/−tax)
Res + NAC +(C/OF +/−tax)
Res only
Mixed
Res + NACRT
(CF, E)
Res + NACRT (CF)
D

B

C
C
C

C
OSA: 0.93 (0.67–1.29)[AA; Caucasian/Chinese/Indian]
OSA: 0.92 (0.63–1.33) [Caucasian]
RecA: 0.40 (0.21–0.78) [Caucasian]
3.06



0.69
1.54
0



0
35
6



4
2
0.660



0.640
0.007
IV rs1801131
(A/C)
MTHFR
Folate metabolism
OSA–Wu 2006 [122] RecA–Wu 2006 [122]

OS and rec–Ott 2011 [98]

OS–Warnecke 2009 [33]
US

AC

SCC
Res + NACRT (CF +/−tax)
Res + NAC (C/OF +/−tax)
Res + NACRT (CF)
D

B

C
OSA: 0.99 (0.64–1.55) [AA; Caucasian]

RecA: 0.80 (0.22–2.95)
0.67



2.38
0



54
3



2
0.970



0.740
IV rs1045642
(C/T)
ABCB1
Drug efflux



OSA and RecA – Wu 2006 [122]
OS – Narumiya 2011 [130]
OS – Okuno 2007 [106]
OS – Warnecke 2009 [33]
US
SCC
US
US
Res + NACRT (CF)
Res + NACRT (CF)
Res + NACRT (CF)
Res + NACRT (CF +/−tax)
D
C
C
D
OSA: 0.57 (0.37–0.87) [TT; Caucasian/Japanese]
OSA: 0.51 (0.32–0.81) [Caucasian]
Rec: 0.26 (0.09–0.81)
1.87

0.68
NA
0

0
NA
4

3
1
0.009

0.004
< 0.05
IV rs11267092
(DEL/INS)
F2R
Angiogensis
OSA and RecA–Lurje 2011 [131] AC Res D OSEx: 1.70 (1.16–2.48)
[INS/INS / INS/DEL; Caucasian]
Rec: 2.41 (1.25–4.65) [INS/INS]
NA NA 1

1
< 0.001

0.003

OS = overall survival; DFS = disease-free survival; rec = recurrence; A = adjusted; (A) = including adjusted; PB = adjusted for publication bias; Ex = extrapolated; NP = not presented; AC = adenocarcinoma; SCC = squamous cell carcinoma; US = unspecified; Res = resection; NAC = neoadjuvant chemotherapy; NACRT = neoadjuvantchemoradiotherapy; DCRT = definitive chemoradiotherapy; C/Cis = cisplatin; O = oxaliplatin; tax = taxane; F = 5FU; E = etoposide; Res = resection; Ex = Extraplotated; INS = insertion; DEL = deletion; NA = not applicable

tumor mutations

Three mutant genes were reported to be associated with outcome: TP53, PIK3CA and NRF2 (LOE IV). TP53 (n = 21) and PIK3CA (n = 3, SCC) underwent meta-analysis.

TP53 status was variably defined and genotyped, although all studies assessed exons 5–8 as a minimum, by single-strand conformation polymorphism (SSCP) analysis with or without sequencing (n = 13), or Sanger sequencing alone (n = 8). Following correction for likely publication bias, a significant negative survival association was demonstrated for mutant TP53 tumors: HR 1.27 (1.01–1.59; P = 0.04; n = 21 studies; supplementary Figures S1 and S2, available at Annals of Oncology online). Significant associations were demonstrated on subgroup meta-analysis of: genotyping technique (SCCP), AC and SCC cell types, and treatment (resection only). Directions of effect were consistent but nonsignificant for adjusted HR alone (n = 6) and use of neoadjuvant chemoradiotherapy (n = 7).

An association with DFS was demonstrated for mutant PIK3CA SCC tumors [HR 0.42 (0.21–0.85); n = 2; P = 0.02], but not OS.

tumor copy number variants

Sixteen tumor CNVs had previously reported associations with prognosis; three were excluded due multiple comparisons (supplementary Table S1, available at Annals of Oncology online). For the remaining 13, LOE was II (1), III (7) and IV (5). Four markers underwent meta-analysis: associations with worse survival were demonstrated for gains in ERBB2 (HER2; LOE III), CCND1 (LOE III) and FGF3 (LOE IV), but not EGFR (LOE II). For all four, there was heterogeneity regarding definition of CNV (absolute copy numbers, or ratio to normal), and genotyping technique [fluorescent/silver in situ hybridization (F/SISH), quantitative (q)PCR and slot/southern blot].

ERBB2/HER2 analysis was restricted to 11 studies performing ISH; 3 (using qPCR [28], or slot [75]/southern [74] blot) were excluded. Worse OS was demonstrated for ERBB2/HER2 gain overall: HR 1.63 (1.20–2.21; P = 2 × 10−4, n = 11). Significance persisted for adjusted HR [2.32 (1.64–2.58); P < 1 × 10−5; n = 3], and cell type (SCC; AC P = 0.06). Treatment regimens were resection alone for all studies, except one [61] including mixed regimens involving the c-MET-GFR inhibitor Crizotinib.

Two meta-analyses were carried out for CCND1: studies using qPCR (n = 4), and slot blot/FISH (n = 2). Worse OS was demonstrated for qPCR [HR = 2.09 (1.27–3.42); P = 0.004], with a concordant nonsignificant trend for FISH/blot. All four studies assessing FGF3 used slot/southern blot; an association with worse OS was demonstrated [HR 1.83 (1.18–2.83); P = 0.006]. EGFR meta-analysis was carried out for FISH and blot techniques (excluding two studies using anti-EGFR therapy [62, 63], and one performing qPCR) [28]. No associations were demonstrated.

loss of heterozygosity

LOH (six markers in total) was associated with outcome by four studies; one study (6p and 13q) was excluded due to multiple comparisons; [132] for another (2p, 3p and 12p), while 3-year survival rates were significant the extrapolated HR was not [86]. 1q22–23 LOH was associated with worse OS in one study (LOE IV).

telomere length ratio

One study reported worse OS with a tumor : normal telomere length >1.17 (LOE III).

genomic instability

CIN was assessed by six studies. Following exclusion of one study including intratumoral heterogeneous ploidy [93] an association with worse OS was demonstrated [HR 1.63 (1.25–2.11); n = 4; P = 0.2 × 10−4]. One study assessed intratumoral heterogeneity alone, reporting better survival than with homogeneity [94]. There were no associations between MSI and survival.

germline polymorphisms

Twenty-nine reported associations were identified (following exclusion of 12 due to multiple comparisons) [107, 109, 112, 133]. Cumulative LOE was II (n = 3), III (n = 22) and IV (n = 4). Fifteen variants underwent meta-analysis (Table 2). Significant associations were demonstrated for six SNPs: ERCC1 rs3212986 (cisplatin treatment; LOE II; Caucasian ethnicity), ERCC2 rs1799793 (cisplatin; Caucasian) TP53 rs1042522 (Caucasian), MDM2 rs2279744 (Caucasian), TYMS rs34743033 (Japanese; LOE III) ABCB1 rs1045642 (Caucasian and Japanese; LOE IV). An association was demonstrated for VEGFA rs2010963, but combining two studies with East Asian ethnicities (Taiwanese and Japanese). One association with recurrence was demonstrated: MTHFR rs1801133 (Caucasian; LOE III).

markers of treatment complications

Twelve reported germline associations (8 studies) were identified; 10 were excluded due to multiple comparisons. One marker (TNFA rs1800629) [134] underwent meta-analysis (nonsignificant). The remaining variant, ACE rs4646994 (LOE III), was associated with postoperative pulmonary complications by one study (Table 3).

Table 3.

Reported tumor variants and germline polymorphisms associated with treatment complications and response to chemo(radio)therapy

LOE Variant Gene Association – mutant Association – wild type No association Cell type Population LOE Meta OR [effect allele / genotype / haplotype] Chi I2 N P
Treatment complications
III rs4646994
(INS/DEL)
ACE
Vasodilator
Post-op pulmonaryA Lee 2005b [135] US Res C ORA: 3.12 (1.01–9.65)
[DEL/DEL; Taiwanese]
NA NA 1 0.049
III rs1800629
(G/A)
TNFA
Cytokine
Post-op infectionA – Azim 2007 [134]
Motoyama 2009 [136]
US
SCC
Res +/− NAC(RT)
Res
C
D
ORA: 4.02 (0.00–18347)
[GG; Caucasian/Japanese]
0 0 2 0.750
Response to chemo(radio)therapy
Tumor
III High DNA ploidy - mPR – HCR versus CR – Ohno 1989 [91] SCC Res + NACRT versus HNACRT (bleomycin) C mPR 13.18 (5.30–32.7) [High ploidy] NA NA 1 < 0.001
IV Mutant (exon) TP53
Apoptosis / DNA repair regulator
mPR – Ribeiro 1998 [43]
mCR –Yamasaki 2010 [41]
cPR – Makino 2010 [47]
mCR – Kunisaki 2006




cCR – Ito 2001 [50]
cCR – Gibson 2003 [55]
US
SCC
SCC
SCC
SCC
US
Res, NACRT (CF + IFN)
Res + NAC
Res + NACRT / DCRT (CF)
DCRT
Res + NACRT (CF)
Res + NACRT (CF)
D
D
D
C
C
C
mPR 0.24 (0.06–0.95) [mutant]
mCR 0.43 (0.08–2.24)

2.87
14.2
30
86
3
3
0.040
0.320
Germline
II rs7121
(T/C)
GNAS1
G protein subunit
mPRA – Alakus 2009 [126] US Res + NACRT (CF) B mPRA 7.25 (1.30–40.62)
[CC; Caucasian]
NA NA 1 < 0.05
III rs3212986
(C/A)
ERCC1
DNA NER
mCRA–Wang 2011 [96]
mPR–Warnecke 2009 [33]
mPR–Rumiato 2013 [97]
SCC
US
US
PC (CF)
Res + NAC (CF)
Res + NAC (CF)
C
B
C
mCR ORA: 2.62 (1.11–6.23)
[AA/CA; Chinese]
mPR OR: 1.52 (0.73–3.20)
[CT/CC; Caucasian]
NA
0.76
NA
0
1
2
< 0.05
0.260
III rs11615
(A/G)
ERCC1
DNA NER
mPR–Metzger 2012 [137]
mPR–Warnecke 2009 [33]


mPR-Rumiato 2013 [97]
AC
US
US
Res + NACRT (CF)
Res + NACRT (CF)
Res + NAC (CF)
D
C
C
mPR OR: 4.57 (3.01–6.94) [TT/CT; Caucasian] 3.48 43 3 < 1x10−5
IV rs25487
(G/C)
XRCC1
DNA repair
cPR–Wu 2006 [122]

cPR - Ott 2011 [98]
mPR–Warnecke 2009 [33]
US
AC
US
Res + NACRT (CF +/−tax)
Res + NAC (C/OF +/−tax))
Res + NACRT(CF)
D
B
C
m/cPR 1.91 (1.30–2.81) [GG; Caucasian] 3.13 36 3 0.001

OS = overall survival; DFS = disease-free survival; rec = recurrence; A = adjusted; AC = adenocarcinoma; SCC = squamous cell carcinoma; US = unspecified; NAC = neoadjuvant chemotherapy; NACRT = neoadjuvantchemoradiotherapy; DCRT = definitive chemoradiotherapy; HNACRT = hyperthermicneoadjuvant chemoradiotherapy; PC = palliative chemotherapy; CF = cisplatin-5FU; DEL = deletion; OF = cisplatin-5FU; Res = resection mPR = major pathological response; cPR = complete pathological response; mCR = major clinical response; cCR = complete clinical response; OR = odds ratio; IFN = interferon; LOE = level of evidence; NA = not applicable

markers of response to chemo(radio)therapy

Two tumor variants (mutant TP53 and CIN) and 15 germline polymorphisms were reported to be associated with clinical or pathological response to chemo ± radiotherapy (Table 3); 11 polymorphisms were excluded due to multiple comparisons).

Mutant TP53 was assessed by six studies; three for pathological and three for clinical response. A lower OR of pathological response was demonstrated [OR 0.24 (0.06–0.95), n = 3, P = 0.04]; effect direction for clinical response was concordant but nonsignificant [OR 0.43 (0.08–2.24); P = 0.32].

Following meta-analysis, two polymorphisms were associated with a major pathological response to platinum-based chemo/radiotherapy in Caucasians: wild-type XRCC1 rs25487 [GG genotype, LOE III; OR 1.91 (1.30–2.81), n = 3, P = 0.001], and variant ERCC1 rs11615 [TT/CT; OR 4.57 (3.01–6.94); n = 3; P < 1 × 10−5]. The AA variant of ERCC1 rs3212986 (LOE III) was associated with radiological response to palliative cisplatin-based chemotherapy in one study (Chinese ethnicity), but not major pathological response to neoadjuvant chemotherapy in two (Caucasian).

markers of stage

Twenty-four tumor markers were reported: 2 mutations, 12 CNV, 7 LOH, 2 MSI and CIN; 15 were excluded.

tumor mutations

Following exclusion of PIK3CA, the sole tumor mutation with a reported association was TP53 (n = 19; LOE III; Table 4). Following meta-analysis, mutant TP53 tumors were associated with more advanced T (T3/T4) and N (≥N1) stages, but not overall TNM stage (III/IV), grade (G3/4) or positive resection margin (R1).

Table 5.

Reported germline markers (polymorphisms) associated with stage of esophageal cancer

LOE Variant Gene Association—variant Association—wild type No association Cell type Population LOE Meta OR [effect allele/genotype/haplotype] Chi I2 N P
II rs6573
(C/A)
RAP1A
RAS oncogene
OA—Wang 2012 [149] SCC Mixed

B OA: 1.89 (1.06–3.36) [CA/AA; Chinese] NA NA 1 0.030
II rs1800471
(G/C)
TGFB1
Growth factor regulator
O, G—Tang 2013 [117] SCC Mixed B O: ORA 2.71 (1.44–5.09) [GC/CC; Chinese]
G: ORA 2.65 (1.44–4.87)
NA

NA
NA

NA
1

1
<0.001

0.002
III rs353163
(T/C)
TMPRSS11A
Serine peptidase
N—Umar 2013b [150] SCC RT/DCRT (CF) C N: 3.27 (1.68–6.39)
[CC; Indian]
NA NA 1 <0.001
III rs2273535
(A/T)
AURKA
Cell cycle kinase
O—Miao 2004 [151] SCC Res C O: 2.13 (1.04–4.39
[TT; Chinese]
NA NA 1 <0.05

III rs7121
(C/T)
GNAS1

G protein subunit
O, N—Vashist 2011 [125] T, M, G—Vashist 2011
T, N, R—Alakus 2009 [126]
US

US
Res

Res + NACRT (CF)
C

B
O: 2.10 (1.17–3.76)

[T; Caucasian]

N: 1.16 (0.76–1.77) [T]
NA



3.94
NA



49
1



3
0.013



0.500

AC, adenocarcinoma; SCC, squamous cell carcinoma; US, unspecified carcinoma; NACRT, neoadjuvant chemoradiotherapy; RT, radiotherapy; DCRT, definitive chemoradiotherapy; T, T stage (III/IV versus I/II); N, nodal stage (≥N1 versus N0); M, metastatic stage (M0 versus M1); G, cell grade (III/IV versus I/II); O, overall stage (III/IV versus I/II); R, resection stage; L, L stage; CF, cisplatin–5FU; Res, resection; LOE, level of evidence; NA, not applicable.

Table 4.

Reported tumor markers (mutations, copy number variants, genomic and chromosomal instability) associated with stage of esophageal cancer

LOE Variant Gene Association – mutant No association Cell type Population LOE Meta OR [effect allele / genotype / haplotype] Chi I2 N P
Mutations
III Mutant TP53
Apoptosis / DNA repair regulator
T, N – Madani 2010 [40]
N – Cao 2004 [54]
N – Hattori 2003 [138]
O Ribeiro 1998








O, T, N, G-Casson 2003 [38]




N - Makino 2010 [47]
R – Madani 2010 [40]
T – Cao 2004 [54]


O, T, N – Schneider 2000 [39]
T, N, G – Soontrapornchai 1999
T, N, G – Egashira 2011 [49]
T, N, M, O – Yamasaki 2010 [41]
T, M, G – Ito 2001 [50]
O – Kobayashi 1999 [42]
T, N, M, G – Uchino 1996 [29]
O – Coggi 1997 [56]

O, T, N – Goan 2005 [53]
O, G – Lam 1997 [51]
O, T, N – Shibagaki 1995 [52]
T, N – Puhringer 2006 [45]
T, M, G – Makino 2010 [47]
AC
SCC
SCC
US
AC
AC
SCC
SCC
SCC
SCC
SCC
US
AC
SCC
SCC
SCC
AC
SCC
Res
Res
Res
Res + NACRT
Res
Mixed
Res
NAC (CF) +/− Res
Res + NACRT (CF)
Res
Res
Res
Res
Res
Res
Res
Res +/− NACRT (CF)
Res +/− NACRT /DCRT (CF)
C
D
C
D
C
D
C
D
C
D
D
D
C
D
D
C
B
D
OPB: 1.28 (0.71–2.31)
T: 1.40 (1.12–1.74)
N: 1.39 (1.07–1.81)
M: 1.21 (0.72–2.03)
G: 1.46 (0.83–2.58)
R: 2.10 (0.470–9.35)

O: B p = 0.540; E p = 0.275
T: B p = 0.393; E p = 0.071
N: B p = 0.765; E p = 0.443
M: B p = 0.207; E = 0.492
G: B p = 0.719; p = 0.543

PB Corrected for Ribeiro 1998
32.4
17.5
18.1
2.99
19.1
3.45
63
20
17
0
53
71
12
15
15
5
10
2
0.410
0.003
0.010
0.480
0.190
0.330
Copy number variants
III Gain SPK2
Protein kinase
N,O – Wang 2009 [139] SCC Res C O: 8.00 (2.25–28.5)
N: 8.10 (2.28–28.8)
NA NA 1
1
1.30x10−3
1.20x10−2
III Gain PRKC1
Serpin
T – Yang 2008 [140] N, O, G – NS – Yang 2008 [140] SCC Res C O: 4.64 (1.71–12.4)
N: 3.12 (1.21–8.02)
T: 2.63 (0.78–8.81)
NA
NA
NA
NA
NA
NA
1
1
1
0.002
0.019
0.118
III Gain HER2 (ERBB2)
Epidermal growth factor receptor
T, G – Yoon 2012 [71]
G,O – Zhan 2012 [68]
O, G – Lennerz 2011 [61]

N – Ikeda 1996 [75]

T, M, L – Zhan 2012 [68]

O, T, N – Suzuki 1997 [74]

O, T, N, M, R, G – Reichelt 2007 [141]
O, N - Brien 2000 [67]

O, N, G - Mimura 2005 [70]
O, T, N, G, R – Sato-Kuwabara 2009 [69]
O, G – Sunpaweravong 2005 [64]
O, T, N, G, M, R – Thompson 2011 [72]
O, T, N, G, M – Al-Kasspooles [142]
T, N, G – Prins 2013 [65]
AC
SCC
US
SCC
SCC
US
AC

SCC
SCC
SCC
AC
AC
AC
Res
Res
Mixed
Res
Res
Res
Res

Res
Res
Res
Res
Res
Res
C
D
D
C
D
C
D

D
D
D
D
C
D
O: 1.13 (0.83–1.54)
T: 0.84 (0.55–1.27)
NPB: 0.96 (0.69–1.35)
M: 1.77 (0.69–4.56)
GPB: 0.61 (0.34–1.09)

PB – Corrected for Ikeda 1996, Al-Kasspooles 1993, Suzuki 1997, Minmura 2005
PB – Corrected for Sunpaweravong 2005, Zhan 2012
14.02
17.5
20.2
3.85
40.0
43
48
31
22
72
10
10
11
4
10
0.510
0.400
0.510
0.240
0.100
IV Gain EGFR
Epidermal growth factor receptor
T,N – Marx 2010 [59]
G – Lennerz 2011 [61]
N – Kitagawa 1996 [60]
N – Yang 2012 [143]
M, G – Marx 2010 [59]
O – Lennerz 2011 [61]
O, T – Kitagawa [60]
O,G – Yang 2012 [143]
O, T,N,M – Miller 2003 [28]
O, T,N,M,G – Al-Kasspooles 1993 [142]
O,T,N,G – Itakura 1994 [31]
AC
US
SCC
SCC
AC
AC
SCC
Res
Mixed
Res
Res
Res
Res
Res
D
D
D
D
C
C
D
OPB: 0.96 (0.6–1.53)
T: 0.51 (0.37–0.71)
N: 0.95 (0.41–2.16)
M: 0.91 (0.29–2.87)
G: 1.24 (0.70–2.20)

PB – Corrected for Kitagawa 1996
9.27
2.31
8.44
1.38
3.36
35
0
41
0
8
6
5
6
3
3
0.850
< 0.001
0.890
0.870
0.460
Loss of heterozygosity
III LOH 3p14.2 TA – Qin 2008 [144] N, M, G – Qin 2008 [144] SCC Res C T: 5.67 (1.77–18.2) NA NA 1 0.003
III LOH 13q T, G – Huang 2002 [145]
N – Harada 1999 [146]
N – Shibagaki 1994 [132]
N – Huang 2002 [145]
T,M,H,O – Harada 1999 [146]
SCC
SCC
SCC
Res
Res
Res
C
C
C
T: 3.08 (0.17–60.8)
N: 4.17 (1.84–9.47)
5.04
3.06
80
35
2
3
0.440
6x10−4
Chromosomal instability
III CIN T,N,G Yu 1989 [147]
G – Doki 1993 [88]
T,N - Kuwano 1995 [92]
N, Ohno 1989 [91]

T,/N – Doki 1993 [88]
N,V,G – Kuwano 1995 [92]
O,T - Ohno 1989 [91]
T,N,G – Tsutsui 1992 [89]
T,L,G – Edwards 1989 [93]
O, T, N G – Wang 1999
T – Kaketani 1989 [90]
SCC
SCC
SCC
SCC
SCC
SCC
SCC
SCC
SCC
Res
Res
Res
Res +/− NACRT
Res
Res
Res +/− AR
Res
Res
C
D
C
C
D
D
D
D
D
O: 2.68 (1.10–6.54)
T: 1.41 (0.97–2.05)
N: 2.18 (1.06–4.47)
G: 1.51 (0.99–2.31)
2.01
7.21
18.2
5.29
0
0
57
5
2
9
7
6
0.030
0.070
0.030
0.060
Microsatellite instability
IV MSI 17q24-25 + Bethesda markers T – Matsumoto 2007 [148] N,G,O – Matsumoto 2007 [148] SCC Res D T: 0.325 (0.11–0.96) NA NA 1 0.043

AC = adenocarcinoma; SCC = squamous cell carcinoma; US = unspecified carcinoma; NAC = neoadjuvant chemotherapy; NACRT = neoadjuvant chemoradiotherapy; CF = cisplatin and 5-fluoruracil; T = T stage (III/IV versus I/II); N = nodal stage(N0 versus ≥N1); M = metastatic stage (M0 versus M1); G = cell grade (III/IV versus I/II); O = overall stage (III/IV versus I/II); R = resection stage (R1 versus R0); L = L stage (L1 versus L0); V = venous invasion (V1 versus V0); Res = resection; PB = corrected for publication bias; LOE = level of evidence; LOH = Loss of Heterozygosity; CIN = chromosomal instability; NA = not applicable

copy number variants

Twelve tumor CNVs were identified; 8 were excluded due to multiple comparisons (Table 4). Meta-analysis was possible for two markers, with one significant association demonstrated: EGFR (T1/2 stage; LOE IV).

loss of heterozygosity

Seven LOH variants were identified; five were excluded (Table 4). Meta-analysis was possible for one marker. A significant association was demonstrated for LOH 13p and ≥N1 stage (Table 4).

genomic instability

Following meta-analysis CIN (LOE III; Table 4) was associated with overall stage [III/IV: OR 2.68 (1.10–6.54); P = 0.03; n = 2 studies] and nodal stage [OR 2.18 (1.06–4.47); P = 0.03; n = 7 studies], but not T stage or grade. MSI (using the 5 Bethesda markers, and 10 at 17q24–25) was associated with more advanced stage; another measure was excluded.

germline polymorphisms

Seventeen polymorphisms were identified; 12 were excluded due to multiple comparisons. One marker (GNAS1 rs7172) underwent meta-analysis, without significance.

funnel plot asymmetry, heterogeneity and publication bias

Begg's and Egger's tests were nonsignificant for all meta-analyses (supplementary Table S26, available at Annals of Oncology online). Visual inspection of plots identified asymmetry for nine outcome analyses: mutant TP53 (OS overall, adjusted HR, SCC and unspecified cell types, neoadjuvant therapy and SSCP/direct sequencing analyses; supplementary Tables S2 and S3, available at Annals of Oncology online), ERRBB2/HER2 (OS) and FGF3 (OS), and three stage analyses: EGFR (overall) and ERBB2/HER2 (N and grade). These were interpreted as likely publication bias and corrected (without affecting any conclusions). All sensitivity analyses were negative.

conclusions

We identified 182 studies, which assessed a total of 165 candidate genomic markers. Overall, 91 markers were reported to have significant associations with esophageal cancer outcome, and 41 with stage. Overall study quality was poor: most studies were retrospective with small sample sizes, and all except 5 (2.75%) were of level C or D quality. There was considerable heterogeneity in patient selection, treatment approach, genotyping techniques and definitions used. Common areas of weakness were failure to perform subgroup analysis for AC and SCC; failure of quality control such as reporting call rates and Hardy–Weinberg equilibrium; failure to perform/report multivariate adjustment of HRs; and failure to adjust for multiple comparisons. Furthermore, just 30.2% of reported markers subsequently had attempted validation data published.

Despite these limitations, sufficient data were available for appropriate meta-analyses. These demonstrated a small number of associations of DNA sequence markers with worse survival (mutant TP53, HER2, CCND1 and FGF3 copy number gain and CIN) and resistance to chemo–radio ± therapy (TP53).

As far as we are aware, this is the first attempt to collate and evaluate all evidence of DNA sequence markers and esophageal cancer, and to demonstrate the above associations by meta-analysis. As such it has a number of generic and specific strengths and weaknesses. A comprehensive search strategy was used to minimize identification and selection bias (requiring detailed appraisal of studies including gastric cancer, cell lines and expression data), it is possible that studies were not identified. For those included, methodological heterogeneity and small sample sizes introduce potential for bias. Although there was no statistical evidence of funnel plot asymmetry using Begg's and Egger's tests, these are underpowered in meta-analyses of fewer than 25 studies [152]; we therefore inspected all funnel plots, explored the reasons for any apparent asymmetry, and corrected eight analyses for likely publication bias (without altering overall effects). While the small number of studies involved in each analysis precluded meaningful meta-regression to explore additional potential confounding factors [153], we sought to address potential bias by performing subgroup analyses, including cell type genotyping techniques and ethnicity. There are also limitations to the revised American Society of Clinical Oncology guidelines in this context; firstly, regarding capture of the complexity inherent in data quality, and secondly determination of LOE: evidence can be upgraded by validation studies, yet disagreement of effect size and direction between studies is not always reflected in the ultimate LOE.

The strongest evidence we found for an outcome marker was tumor TP53 mutation. Association with worse survival was demonstrated for both AC and SCC. Whether this is truly independent of the association demonstrated with T and N stage (independent pathological markers of outcome) [154] was not conclusively demonstrated, and indeed only assessed by six studies. Although four reported significant adjusted HR, the resultant meta-analyzed direction of effect was concordant but not significant due to the use of a random-effects model. We also found TP53 mutant tumors to be less chemo(radio)sensitive.

As other recent meta-analyses have reported similar findings in breast and colorectal carcinoma [155, 156], this is of particular translatable relevance. TP53 is one of the most frequently mutated and studied genes in human cancer [157], with resultant attempts to develop targeted therapies [158]. Ninety-five percent of functional mutations occur within exons 5–9, which encode the DNA binding domain, and typically cause loss of efficacy either directly by disrupting DNA contact, or indirectly by aberrant protein folding [159]. Subsequently, cell cycle, DNA repair and apoptotic regulation may fail [160], although oncogenic gains of function are occasionally seen [161]. The most characterized variant is the germline rs1042522 G > C substitution, itself conferring a worse HR for both OS and DFS in this meta-analysis.

TP53 as an esophageal tumor biomarker is often considered in terms of TP53 status: aberrant expression, with or without mutation. An association with expression alone and worse outcome has been demonstrated on meta-analysis for SCC [162], as has aberrant status [increased expression (28 studies) with or without mutation (3 studies)] and reduced likelihood of response to chemotherapy [163]. However, TP53 mutational and expression statuses may be discordant [164] particularly in the case of high-impact mutations precluding expression, or dramatically reducing half-life. The ability to predict this from sequencing data reinforces the need to explore the interaction of these aspects of status in parallel [165, 166].

Typically, resection specimens are used to assess associations between tumor markers and pathological response to chemotherapy. However, by definition these comprise clonal populations selected for chemo/radio-resistance. While such tumors appear to be disproportionately TP53 mutated, deep re-sequencing and clonal studies comparing the prevalence and associations of pre- and post-treatment tumor are required to establish the true pretreatment predictive utility of TP53 mutations in this regard.

Three associations between tumor copy number gain (albeit variably quantified) were demonstrated by meta-analysis: ERBB2/HER2, CCND1 and FGF3 gain. ERBB2/HER2 is particularly relevant; a proto-oncogene, it is the sole molecular marker in clinical use for gastroesophageal cancer, guiding the use of targeted therapies [14]. Our findings build on a recent meta-analysis of HER2 status, defining positivity by overexpression or amplification, including six of the studies included in this meta-analysis [15]. We found gain to confer a worse prognosis for both AC and SCC, independent of stage. Interestingly, all patients in 10 of the 11 studies underwent radical treatment with resection; while palliative monoclonal antibody therapy for HER2-positive gastroesophageal AC is effective in prolonging survival [167], an urgent unanswered question is therefore whether it has a role in curative treatment.

Similarly, regarding the cell cycle regulator CCND1, phase I and II data have suggested a possible role for cyclin-dependent kinase inhibitors in nongastroesophgageal cancer [168, 169]. Our findings therefore suggest the need to assess their effect in esophageal tumors with CCND1 gain. EGFR, a tyrosine kinase receptor, has also been extensively investigated within gastroesophageal cancer; phase II data support targeted therapy (antibodies and tyrosine kinase inhibitors) for metastatic disease [170, 171], although not yet neoadjuvant regimens [172, 173]. While we found no association with outcome using the requisite random-effects model, significant effects were evident with a fixed model; consequently, there may be an undetected association. We also found CIN to be associated with worse outcome, in keeping with a previous colorectal cancer meta-analysis [174], although whether it modulates chemo-sensitivity is unclear.

We also demonstrated survival associations for six common germline polymorphisms by meta-analysis: ERCC1 rs3212986 (for cisplatin treatment and Caucasian ethnicity), ERCC2 rs1799793 (cisplatin and Caucasian), TP53 rs1042522 (Caucasian), MDM2 rs2279744 (Caucasian), TYMS rs34743033 (Japanese) ABCB1 rs1045642 (both Caucasian and Japanese). The association of VEGFA rs2010963 was evident only on combining Taiwanese and Japanese study populations. MTHFR rs1801133 was associated with recurrence in Caucasians. XRCC1 rs25487 and ERCC1 rs11615 were associated with response to chemotherapy in Caucasians. These associations are likely to be due to aberrant protein expression or function.

rs3212986 modifies ERCC1 mRNA stability [175], a component of the nucleotide excision repair (NER) pathway, variants of which are associated with platinum sensitivity and survival in pancreatic, gastric, colorectal and lung cancers [95177]. The missense rs1799793 SNP results in an aspartate–asparagine substitution at codon 312 of the ERCC2 component of the NER pathway, and has been similarly associated with survival in gastric and other cancers [178]. The rs10456402 SNP in exon 26 of ABCB1 (Multi Drug Resistance 1) reduces expression (and consequent platinum-analogue membrane transportation) [179], and is similarly associated with colorectal cancer prognosis [180]. rs2279744 increases mRNA expression of MDM2, which suppresses TP53 activity [181], and is associated with increased susceptibility to a number of cancers (including gastric) [182]. rs34743033 is a 28-bp variable number tandem repeat in TYMS (thymidylate synthase), with enhancer function correlating with increased TYMS expression [183], and survival in platinum-treatment nonsmall-cell lung carcinoma [184]. The rs1801133 missense SNP induces an alanine–valine substitution at codon 222, with reduced activity of methylenetetrahydrofolate reductase [185], and increased susceptibility to gastric cancer [186]. rs25487 induces a glutamine-arginine substation in codon 399, with resultant reduction in function of the DNA repair gene XRCC1 [187], and an association with survival of lung cancer [188]. rs11615 reduces ERCC1 expression [189], and increases likelihood of response to platinum chemotherapy in gastric and colorectal cancer [176].

Biomarkers themselves carry a number of limitations. Typically, they are classified as ‘prognostic’ or ‘predictive’; however, in reality, these are not mutually exclusive, and we therefore did not attempt classification. Biomarker development culminates in demonstration of clinical, requiring at least multicenter prospective validation for prognosis, and incorporation into interaction randomized controlled trials for prediction. These challenges reinforce the utility of retrospectively analyzing samples archived during prospective trials. Other challenges include the use of pretreatment biopsies. First, analysis may be impaired by inclusion of noncancerous tissue; while this can be mitigated by techniques such as laser-capture microdissection and higher depth sequencing, these are time and cost-intensive. More profound is the challenge of intratumoral heterogeneity and clonality: a single biopsy is representative of just 34% of the mutational burden of a ‘single’ cancer [190], and will not include metastatic subclones. How to surmount this is not yet clear.

Finally, while it may be pragmatic to consider DNA sequence variants in isolation, their effects (and therefore utility) are subject to complicated modulation by the other ‘omics', (epigenomics, transcriptomics, metabolomics and proteomics), genes and clinical and environmental covariates [191, 192]. While a discrete variable might provide useful complementary information of itself, this complexity at present precludes its use to dichotomize decision making. Consequently, a robust approach to personalized cancer medicine must incorporate parallel processing of DNA, RNA, proteins and metabolites.

In conclusion, numerous DNA sequence markers have been described for esophageal cancer. However, as with complementary fields within personalized cancer research, the underlying research is largely poor in quality and disparate in methodology, with a lack of robust validation of markers and incorporation into trials. While a number of promising candidates have been identified the data required to incorporate these into prognostic/predictive models do not yet exist; future validation will require larger studies, with improvements in the standardized collection of samples for analysis, parallel assessment of expression and the incorporation of parallel biomarkers within high-quality clinical trials, robust adjustment for confounding variables and sharing of resultant data with multicenter collaboration.

funding

JF is supported by the NIHR Oxford Biomedical Research Centre. Core funding is provided to the Wellcome Trust Centre for Human Genetics from the Wellcome Trust (090532/Z/09/Z).

disclosure

MRM has received payment for advisory/consulting roles within the last 2 years from Amgen, Bristol-Meyers Sqibb, GlaxoSmithKline, Merck, Millennium, and Roche, and institutional funding from Amgen, AstraZeneca, Bristol-Meyers Squibb, Clovis, Eisai, GlaxoSmithKline, Immunocore, Johnson & Johnson, Merck, Millennium, Novartis, Pfizer, Roche and Vertex. The authors have declared no conflicts of interest.

Supplementary Material

Supplementary Data

references

  • 1.Ferlay J, Parkin DM, Steliarova-Foucher E. Estimates of cancer incidence and mortality in Europe in 2008. Eur J Cancer 2010; 46: 765–781. [DOI] [PubMed] [Google Scholar]
  • 2.Lepage C, Rachet B, Jooste V, et al. Continuing rapid increase in esophageal adenocarcinoma in England and Wales. Am J Gastroenterol 2008; 103: 2694–2699. [DOI] [PubMed] [Google Scholar]
  • 3.Lagergren J, Bergstrom R, Lindgren A, et al. Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N Engl J Med 1999; 340: 825–831. [DOI] [PubMed] [Google Scholar]
  • 4.Rodgers M, Jobe BA, O'Rourke RW, et al. Case volume as a predictor of inpatient mortality after esophagectomy. Arch Surg 2007; 142: 829–839. [DOI] [PubMed] [Google Scholar]
  • 5.Unit RCoSCE National Oesophago-Gastric Cancer Audit 2012. London: Royal College Of Surgeons of England 2012.
  • 6.Findlay JM, Tustian E, Millo J, et al. The effect of formalizing enhanced recovery after esophagectomy with a protocol. Dis Esophagus 2014. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 7.Scarpa M, Saadeh LM, Fasolo A, et al. Health-related quality of life in patients with oesophageal cancer: analysis at different steps of the treatment pathway. J Gastrointest Surg 2013; 17: 421–433. [DOI] [PubMed] [Google Scholar]
  • 8.Djarv T, Derogar M, Lagergren P. Influence of co-morbidity on long-term quality of life after oesophagectomy for cancer. Br J Surg 2014; 101: 495–501. [DOI] [PubMed] [Google Scholar]
  • 9.Gebski V, Burmeister B, Smithers BM, et al. Survival benefits from neoadjuvant chemoradiotherapy or chemotherapy in oesophageal carcinoma: a meta-analysis. Lancet Oncol 2007; 8: 226–234. [DOI] [PubMed] [Google Scholar]
  • 10.Campbell NP, Villaflor VM. Neoadjuvant treatment of esophageal cancer. World J Gastroenterol 2010; 16: 3793–3803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Rouvelas I, Zeng W, Lindblad M, et al. Survival after surgery for oesophageal cancer: a population-based study. Lancet Oncol 2005; 6: 864–870. [DOI] [PubMed] [Google Scholar]
  • 12.Jamieson GG, Mathew G, Ludemann R, et al. Postoperative mortality following oesophagectomy and problems in reporting its rate. Br J Surg 2004; 91: 943–947. [DOI] [PubMed] [Google Scholar]
  • 13.Davies AR, Pillai A, Sinha P, et al. Factors associated with early recurrence and death after esophagectomy for cancer. J Surg Oncol 2014; 109: 459–464. [DOI] [PubMed] [Google Scholar]
  • 14.Allum WH, Blazeby JM, Griffin SM, et al. Guidelines for the management of oesophageal and gastric cancer. Gut 2011; 60: 1449–1472. [DOI] [PubMed] [Google Scholar]
  • 15.Chan DS, Twine CP, Lewis WG. Systematic review and meta-analysis of the influence of HER2 expression and amplification in operable oesophageal cancer. J Gastrointest Surg 2012; 16: 1821–1829. [DOI] [PubMed] [Google Scholar]
  • 16.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Agrawal N, Jiao Y, Bettegowda C, et al. Comparative genomic analysis of esophageal adenocarcinoma and squamous cell carcinoma. Cancer Discov 2012; 2: 899–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kim JC, Kim SY, Cho DH, et al. Novel chemosensitive single-nucleotide polymorphism markers to targeted regimens in metastatic colorectal cancer. Clin Cancer Res 2011; 17: 1200–1209. [DOI] [PubMed] [Google Scholar]
  • 20.Patel JN, McLeod HL, Innocenti F. Implications of genome-wide association studies in cancer therapeutics. Br J Clin Pharmacol 2013; 76: 370–380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rice TW, Blackstone EH, Rusch VW. 7th edition of the AJCC Cancer Staging Manual: esophagus and esophagogastric junction. Ann Surg Oncol 2010; 17: 1721–1724. [DOI] [PubMed] [Google Scholar]
  • 22.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. [DOI] [PubMed] [Google Scholar]
  • 23.Gray KA, Daugherty LC, Gordon SM, et al. Genenames.org: the HGNC resources in 2013. Nucleic Acids Res 2013; 41: D545–D552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bland JM, Altman DG. Multiple significance tests: the Bonferroni method. BMJ 1995; 310: 170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.International HapMap C. The International HapMap Project. Nature 2003; 426: 789–796. [DOI] [PubMed] [Google Scholar]
  • 27.Parmar MK, Torri V, Stewart L. Extracting summary statistics to perform meta-analyses of the published literature for survival endpoints. Stat Med 1998; 17: 2815–2834. [DOI] [PubMed] [Google Scholar]
  • 28.Miller CT, Moy JR, Lin L, et al. Gene amplification in esophageal adenocarcinomas and Barrett's with high-grade dysplasia. Clin Cancer Res 2003; 9: 4819–4825. [PubMed] [Google Scholar]
  • 29.Uchino S, Saito T, Inomata M, et al. Prognostic significance of the p53 mutation in esophageal cancer. Jpn J Clin Oncol 1996; 26: 287–292. [DOI] [PubMed] [Google Scholar]
  • 30.Chikuba K, Saito T, Uchino S, et al. High amplification of the hst-1 gene correlates with haematogenous recurrence after curative resection of oesophageal carcinoma. Br J Surg 1995; 82: 364–367. [DOI] [PubMed] [Google Scholar]
  • 31.Itakura Y, Sasano H, Shiga C, et al. Epidermal growth factor receptor overexpression in esophageal carcinoma. An immunohistochemical study correlated with clinicopathologic findings and DNA amplification. Cancer 1994; 74: 795–804. [DOI] [PubMed] [Google Scholar]
  • 32.Wang LS, Chow KC, Chi KH, et al. Prognosis of esophageal squamous cell carcinoma: analysis of clinicopathological and biological factors. Am J Gastroenterol 1999; 94: 1933–1940. [DOI] [PubMed] [Google Scholar]
  • 33.Warnecke-Eberz U, Vallbohmer D, Alakus H, et al. ERCC1 and XRCC1 gene polymorphisms predict response to neoadjuvant radiochemotherapy in esophageal cancer. J Gastrointest Surg 2009; 13: 1411–1421. [DOI] [PubMed] [Google Scholar]
  • 34.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. [DOI] [PubMed] [Google Scholar]
  • 35.Higgins J, Green SP. Cochrane Handbook for Systematic Reviews of Interventions. Oxford: Wiley-Blackwell 2008. [Google Scholar]
  • 36.Sutton AJ, Duval SJ, Tweedie RL, et al. Empirical assessment of effect of publication bias on meta-analyses. BMJ 2000; 320: 1574–1577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Development Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing: 2013. [Google Scholar]
  • 38.Casson AG, Evans SC, Gillis A, et al. Clinical implications of p53 tumor suppressor gene mutation and protein expression in esophageal adenocarcinomas: results of a ten-year prospective study. J Thorac Cardiovasc Surg 2003; 125: 1121–1131. [DOI] [PubMed] [Google Scholar]
  • 39.Schneider PM, Stoeltzing O, Roth JA, et al. P53 mutational status improves estimation of prognosis in patients with curatively resected adenocarcinoma in Barrett's esophagus. Clin Cancer Res 2000; 6: 3153–3158. [PubMed] [Google Scholar]
  • 40.Madani K, Zhao R, Lim HJ, et al. Prognostic value of p53 mutations in oesophageal adenocarcinoma: final results of a 15-year prospective study. Eur J Cardiothorac Surg 2010; 37: 1427–1432. [DOI] [PubMed] [Google Scholar]
  • 41.Yamasaki M, Miyata H, Fujiwara Y, et al. p53 genotype predicts response to chemotherapy in patients with squamous cell carcinoma of the esophagus. Ann Surg Oncol 2010; 17: 634–642. [DOI] [PubMed] [Google Scholar]
  • 42.Kobayashi S, Koide Y, Endo M, et al. The p53 gene mutation is of prognostic value in esophageal squamous cell carcinoma patients in unified stages of curability. Am J Surg 1999; 177: 497–502. [DOI] [PubMed] [Google Scholar]
  • 43.Ribeiro U, Jr, Finkelstein SD, Safatle-Ribeiro AV, et al. p53 sequence analysis predicts treatment response and outcome of patients with esophageal carcinoma. Cancer 1998; 83: 7–18. [PubMed] [Google Scholar]
  • 44.Kandioler D, Schoppmann SF, Zwrtek R, et al. The biomarker TP53 divides patients with neoadjuvantly treated esophageal cancer into 2 subgroups with markedly different outcomes. A p53 Research Group study. J Thorac Cardiovasc Surg 2014. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 45.Puhringer-Oppermann F, Stahl M, Keller G, et al. Lack of prognostic impact of p53 gene mutation and p53 phosphorylation at serine 15 in multimodally treated adenocarcinomas of the gastroesophageal junction. J Cancer Res Clin Oncol 2006; 132: 433–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Soontrapornchai P, Elsaleh H, Joseph D, et al. TP53 gene mutation status in pretreatment biopsies of oesophageal adenocarcinoma has no prognostic value. Eur J Cancer 1999; 35: 1683–1687. [DOI] [PubMed] [Google Scholar]
  • 47.Makino T, Yamasaki M, Miyata H, et al. p53 Mutation status predicts pathological response to chemoradiotherapy in locally advanced esophageal cancer. Ann Surg Oncol 2010; 17: 804–811. [DOI] [PubMed] [Google Scholar]
  • 48.Shimada Y, Imamura M, Shibagaki I, et al. Genetic alterations in patients with esophageal cancer with short- and long-term survival rates after curative esophagectomy. Ann Surg 1997; 226: 162–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Egashira A, Morita M, Yoshida R, et al. Loss of p53 in esophageal squamous cell carcinoma and the correlation with survival: analyses of gene mutations, protein expression, and loss of heterozygosity in Japanese patients. J Surg Oncol 2011; 104: 169–175. [DOI] [PubMed] [Google Scholar]
  • 50.Ito T, Kaneko K, Makino R, et al. Prognostic value of p53 mutations in patients with locally advanced esophageal carcinoma treated with definitive chemoradiotherapy. J Gastroenterol 2001; 36: 303–311. [DOI] [PubMed] [Google Scholar]
  • 51.Lam KY, Tsao SW, Zhang D, et al. Prevalence and predictive value of p53 mutation in patients with oesophageal squamous cell carcinomas: a prospective clinico-pathological study and survival analysis of 70 patients. Int J Cancer 1997; 74: 212–219. [DOI] [PubMed] [Google Scholar]
  • 52.Shibagaki I, Tanaka H, Shimada Y, et al. p53 mutation, murine double minute 2 amplification, and human papillomavirus infection are frequently involved but not associated with each other in esophageal squamous cell carcinoma. Clin Cancer Res 1995; 1: 769–773. [PubMed] [Google Scholar]
  • 53.Goan YG, Hsu HK, Chang HC, et al. Deregulated p21(WAF1) overexpression impacts survival of surgically resected esophageal squamous cell carcinoma patients. Ann Thorac Surg 2005; 80: 1007–1016. [DOI] [PubMed] [Google Scholar]
  • 54.Cao W, Chen X, Dai H, et al. Mutational spectra of p53 in geographically localized esophageal squamous cell carcinoma groups in China. Cancer 2004; 101: 834–844. [DOI] [PubMed] [Google Scholar]
  • 55.Gibson MK, Abraham SC, Wu TT, et al. Epidermal growth factor receptor, p53 mutation, and pathological response predict survival in patients with locally advanced esophageal cancer treated with preoperative chemoradiotherapy. Clin Cancer Res 2003; 9: 6461–6468. [PubMed] [Google Scholar]
  • 56.Coggi G, Bosari S, Roncalli M, et al. p53 protein accumulation and p53 gene mutation in esophageal carcinoma. A molecular and immunohistochemical study with clinicopathologic correlations. Cancer 1997; 79: 425–432. [DOI] [PubMed] [Google Scholar]
  • 57.Izzo JG, Wu TT, Wu X, et al. Cyclin D1 guanine/adenine 870 polymorphism with altered protein expression is associated with genomic instability and aggressive clinical biology of esophageal adenocarcinoma. J Clin Oncol 2007; 25: 698–707. [DOI] [PubMed] [Google Scholar]
  • 58.Shigaki H, Baba Y, Watanabe M, et al. PIK3CA mutation is associated with a favorable prognosis among patients with curatively resected esophageal squamous cell carcinoma. Clin Cancer Res 2013; 19: 2451–2459. [DOI] [PubMed] [Google Scholar]
  • 59.Marx AH, Zielinski M, Kowitz CM, et al. Homogeneous EGFR amplification defines a subset of aggressive Barrett's adenocarcinomas with poor prognosis. Histopathology 2010; 57: 418–426. [DOI] [PubMed] [Google Scholar]
  • 60.Kitagawa Y, Ueda M, Ando N, et al. Further evidence for prognostic significance of epidermal growth factor receptor gene amplification in patients with esophageal squamous cell carcinoma. Clin Cancer Res 1996; 2: 909–914. [PubMed] [Google Scholar]
  • 61.Lennerz JK, Kwak EL, Ackerman A, et al. MET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinib. J Clin Oncol 2011; 29: 4803–4810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Luber B, Deplazes J, Keller G, et al. Biomarker analysis of cetuximab plus oxaliplatin/leucovorin/5-fluorouracil in first-line metastatic gastric and oesophago-gastric junction cancer: results from a phase II trial of the Arbeitsgemeinschaft Internistische Onkologie (AIO). BMC Cancer 2011; 11: 509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Janmaat ML, Gallegos-Ruiz MI, Rodriguez JA, et al. Predictive factors for outcome in a phase II study of gefitinib in second-line treatment of advanced esophageal cancer patients. J Clin Oncol 2006; 24: 1612–1619. [DOI] [PubMed] [Google Scholar]
  • 64.Sunpaweravong P, Sunpaweravong S, Puttawibul P, et al. Epidermal growth factor receptor and cyclin D1 are independently amplified and overexpressed in esophageal squamous cell carcinoma. J Cancer Res Clin Oncol 2005; 131: 111–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Prins MJ, Ruurda JP, van Diest PJ, et al. The significance of the HER-2 status in esophageal adenocarcinoma for survival: an immunohistochemical and an in situ hybridization study. Ann Oncol 2013; 24: 1290–1297. [DOI] [PubMed] [Google Scholar]
  • 66.Rauser S, Weis R, Braselmann H, et al. Significance of HER2 low-level copy gain in Barrett's cancer: implications for fluorescence in situ hybridization testing in tissues. Clin Cancer Res 2007; 13: 5115–5123. [DOI] [PubMed] [Google Scholar]
  • 67.Brien TP, Odze RD, Sheehan CE, et al. HER-2/neu gene amplification by FISH predicts poor survival in Barrett's esophagus-associated adenocarcinoma. Hum Pathol 2000; 31: 35–39. [DOI] [PubMed] [Google Scholar]
  • 68.Zhan N, Dong WG, Tang YF, et al. Analysis of HER2 gene amplification and protein expression in esophageal squamous cell carcinoma. Med Oncol 2012; 29: 933–940. [DOI] [PubMed] [Google Scholar]
  • 69.Sato-Kuwabara Y, Neves JI, Fregnani JH, et al. Evaluation of gene amplification and protein expression of HER-2/neu in esophageal squamous cell carcinoma using Fluorescence in situ Hybridization (FISH) and immunohistochemistry. BMC Cancer 2009; 9: 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Mimura K, Kono K, Hanawa M, et al. Frequencies of HER-2/neu expression and gene amplification in patients with oesophageal squamous cell carcinoma. Br J Cancer 2005; 92: 1253–1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Yoon HH, Shi Q, Sukov WR, et al. Adverse prognostic impact of intratumor heterogeneous HER2 gene amplification in patients with esophageal adenocarcinoma. J Clin Oncol 2012; 30: 3932–3938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Thompson SK, Sullivan TR, Davies R, et al. Her-2/neu gene amplification in esophageal adenocarcinoma and its influence on survival. Ann Surg Oncol 2011; 18: 2010–2017. [DOI] [PubMed] [Google Scholar]
  • 73.Konig AM, Reeh M, Dancau AM, et al. Concordance of HER2 status in primary tumour and lymph node metastases in patients with esophageal carcinoma. Anticancer Res 2013; 33: 4975–4982. [PubMed] [Google Scholar]
  • 74.Suzuki H, Abo S, Kitamura M, et al. Gene amplification of int-2 and erbB in human esophageal cancer: relationship to clinicopathological variables. Cancer Invest 1997; 15: 411–415. [DOI] [PubMed] [Google Scholar]
  • 75.Ikeda Y, Ozawa S, Ando N, et al. Meanings of c-erbB and int-2 amplification in superficial esophageal squamous cell carcinomas. Ann Thorac Surg 1996; 62: 835–838. [DOI] [PubMed] [Google Scholar]
  • 76.Wang MT, Chen G, An SJ, et al. Prognostic significance of cyclinD1 amplification and the co-alteration of cyclinD1/pRb/ppRb in patients with esophageal squamous cell carcinoma. Dis Esophagus 2012; 25: 664–670. [DOI] [PubMed] [Google Scholar]
  • 77.Takeshita H, Ichikawa D, Komatsu S, et al. Prediction of CCND1 amplification using plasma DNA as a prognostic marker in oesophageal squamous cell carcinoma. Br J Cancer 2010; 102: 1378–1383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Shinozaki H, Ozawa S, Ando N, et al. Cyclin D1 amplification as a new predictive classification for squamous cell carcinoma of the esophagus, adding gene information. Clin Cancer Res 1996; 2: 1155–1161. [PubMed] [Google Scholar]
  • 79.Komatsu S, Ichikawa D, Hirajima S, et al. Clinical impact of predicting CCND1 amplification using plasma DNA in superficial esophageal squamous cell carcinoma. Dig Dis Sci 2014; 59: 1152–1159. [DOI] [PubMed] [Google Scholar]
  • 80.Gramlich TL, Fritsch CR, Maurer D, et al. Differential polymerase chain reaction assay of cyclin D1 gene amplification in esophageal carcinoma. Diagn Mol Pathol 1994; 3: 255–259. [DOI] [PubMed] [Google Scholar]
  • 81.Carneiro A, Isinger A, Karlsson A, et al. Prognostic impact of array-based genomic profiles in esophageal squamous cell cancer. BMC Cancer 2008; 8: 98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Mori M, Tokino T, Yanagisawa A, et al. Association between chromosome 11q13 amplification and prognosis of patients with oesophageal carcinomas. Eur J Cancer 1992; 28A: 755–757. [DOI] [PubMed] [Google Scholar]
  • 83.Wang YF, Wang XS, Gao SG, et al. Clinical significance of combined detection of human papilloma virus infection and human telomerase RNA component gene amplification in patients with squamous cell carcinoma of the esophagus in northern China. Eur J Med Res 2013; 18: 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Shi ZZ, Liang JW, Zhan T, et al. Genomic alterations with impact on survival in esophageal squamous cell carcinoma identified by array comparative genomic hybridization. Genes Chromosomes Cancer 2011; 50: 518–526. [DOI] [PubMed] [Google Scholar]
  • 85.Gertler R, Doll D, Maak M, et al. Telomere length and telomerase subunits as diagnostic and prognostic biomarkers in Barrett carcinoma. Cancer 2008; 112: 2173–2180. [DOI] [PubMed] [Google Scholar]
  • 86.Ikeguchi M, Unate H, Maeta M, et al. Detection of loss of heterozygosityat microsatellite loci in esophageal squamous-cell carcinoma. Oncology 1999; 56: 164–168. [DOI] [PubMed] [Google Scholar]
  • 87.Maru DM, Luthra R, Correa AM, et al. Frequent loss of heterozygosity of chromosome 1q in esophageal adenocarcinoma: loss of chromosome 1q21.3 is associated with shorter overall survival. Cancer 2009; 115: 1576–1585. [DOI] [PubMed] [Google Scholar]
  • 88.Doki Y, Shiozaki H, Tahara H, et al. Prognostic value of DNA ploidy in squamous cell carcinoma of esophagus. Analyzed with improved flow cytometric measurement. Cancer 1993; 72: 1813–1818. [DOI] [PubMed] [Google Scholar]
  • 89.Tsutsui S, Kuwano H, Mori M, et al. A flow cytometric analysis of DNA content in primary and metastatic lesions of esophageal squamous cell carcinoma. Cancer 1992; 70: 2586–2591. [DOI] [PubMed] [Google Scholar]
  • 90.Kaketani K, Saito T, Kobayashi M. Flow cytometric analysis of nuclear DNA content in esophageal cancer. Aneuploidy as an index for highly malignant potential. Cancer 1989; 64: 887–891. [DOI] [PubMed] [Google Scholar]
  • 91.Ohno S, Korenaga D, Kuwano H, et al. DNA aneuploidy assessment of the effectiveness of hyperthermo-chemo-radiotherapy for esophageal carcinoma. Cancer 1989; 63: 1951–1955. [DOI] [PubMed] [Google Scholar]
  • 92.Kuwano H, Sumiyoshi K, Nozoe T, et al. The prognostic significance of the cytophotometric DNA content and its relationship with the argyrophilic nucleolar organizer regions (AgNOR) and proliferating cell nuclear antigen (PCNA) in oesophageal cancer. Eur J Surg Oncol 1995; 21: 368–373. [DOI] [PubMed] [Google Scholar]
  • 93.Edwards JM, Jones DJ, Wilkes SJ, et al. Ploidy as a prognostic indicator in oesophageal squamous carcinoma and its relationship to various histological criteria. J Pathol 1989; 159: 35–41. [DOI] [PubMed] [Google Scholar]
  • 94.Deguchi S, Muto Y, Kusano T, et al. Intratumoral heterogeneity of DNA ploidy and regional differences in epidermal growth factor and epidermal growth factor receptor of esophageal carcinoma. Tohoku J Exp Med 1993; 171: 107–118. [DOI] [PubMed] [Google Scholar]
  • 95.Bradbury PA, Kulke MH, Heist RS, et al. Cisplatin pharmacogenetics, DNA repair polymorphisms, and esophageal cancer outcomes. Pharmacogenet Genomics 2009; 19: 613–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Wang Y, Chen J, Li X, et al. Genetic polymorphisms of ERCC1 and their effects on the efficacy of cisplatin-based chemotherapy in advanced esophageal carcinoma. Oncol Rep 2011; 25: 1047–1052. [DOI] [PubMed] [Google Scholar]
  • 97.Rumiato E, Cavallin F, Boldrin E, et al. ERCC1 C8092A (rs3212986) polymorphism as a predictive marker in esophageal cancer patients treated with cisplatin/5-FU-based neoadjuvant therapy. Pharmacogenet Genomics 2013; 23: 597–604. [DOI] [PubMed] [Google Scholar]
  • 98.Ott K, Rachakonda PS, Panzram B, et al. DNA repair gene and MTHFR gene polymorphisms as prognostic markers in locally advanced adenocarcinoma of the esophagus or stomach treated with cisplatin and 5-fluorouracil-based neoadjuvant chemotherapy. Ann Surg Oncol 2011; 18: 2688–2698. [DOI] [PubMed] [Google Scholar]
  • 99.Wu C, Li M, Hu C, et al. Prognostic role of microRNA polymorphisms in patients with advanced esophageal squamous cell carcinoma receiving platinum-based chemotherapy. Cancer Chemother Pharmacol 2014; 73: 335–341. [DOI] [PubMed] [Google Scholar]
  • 100.Yang PW, Huang YC, Hsieh CY, et al. Association of miRNA-related genetic polymorphisms and prognosis in patients with esophageal squamous cell carcinoma. Ann Surg Oncol, 2014. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 101.Vashist YK, Trump F, Gebauer F, et al. EGFR intron-1 CA repeat polymorphism is a predictor of relapse and survival in complete resected only surgically treated esophageal cancer. Target Oncol 2014; 9: 43–52. [DOI] [PubMed] [Google Scholar]
  • 102.Motoyama S, Nakatsu T, Miura M, et al. Interleukin-6–634G>C genetic polymorphism is associated with prognosis following surgery for advanced thoracic esophageal squamous cell carcinoma. Dig Surg 2012; 29: 194–201. [DOI] [PubMed] [Google Scholar]
  • 103.Lee JM, Yang PW, Yang SY, et al. Genetic variants in DNA repair predicts the survival of patients with esophageal cancer. Ann Surg 2011; 253: 918–927. [DOI] [PubMed] [Google Scholar]
  • 104.Yang PW, Hsieh CY, Kuo FT, et al. The survival impact of XPA and XPC genetic polymorphisms on patients with esophageal squamous cell carcinoma. Ann Surg Oncol 2013; 20: 562–571. [DOI] [PubMed] [Google Scholar]
  • 105.Kaneko K, Nagai M, Murakami Y, et al. TS gene tandem repeats in esophageal cancer patients receiving chemoradiotherapy. Front Biosci 2011; 16: 1036–1043. [DOI] [PubMed] [Google Scholar]
  • 106.Okuno T, Tamura T, Yamamori M, et al. Favorable genetic polymorphisms predictive of clinical outcome of chemoradiotherapy for stage II/III esophageal squamous cell carcinoma in Japanese. Am J Clin Oncol 2007; 30: 252–257. [DOI] [PubMed] [Google Scholar]
  • 107.Sarbia M, Stahl M, von Weyhern C, et al. The prognostic significance of genetic polymorphisms (Methylenetetrahydrofolate Reductase C677T, Methionine Synthase A2756G, Thymidilate Synthase tandem repeat polymorphism) in multimodally treated oesophageal squamous cell carcinoma. Br J Cancer 2006; 94: 203–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Renouf DJ, Zhai R, Sun B, et al. Association of MDM2 T309G and p53 Arg72Pro polymorphisms and gastroesophageal reflux disease with survival in esophageal adenocarcinoma. J Gastroenterol Hepatol 2013; 28: 1482–1488. [DOI] [PubMed] [Google Scholar]
  • 109.Boonstra JJ, van Marion R, Tilanus HW, et al. Functional polymorphisms associated with disease-free survival in resected carcinoma of the esophagus. J Gastrointest Surg 2011; 15: 48–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Cescon DW, Bradbury PA, Asomaning K, et al. p53 Arg72Pro and MDM2 T309G polymorphisms, histology, and esophageal cancer prognosis. Clin Cancer Res 2009; 15: 3103–3109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Pan JY, Ajani JA, Gu J, et al. Association of Aurora-A (STK15) kinase polymorphisms with clinical outcome of esophageal cancer treated with preoperative chemoradiation. Cancer 2012; 118: 4346–4353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Tamura T, Kuwahara A, Yamamori M, et al. VEGF -634C/G genotype is predictive of long-term survival after treatment with a definitive 5-fluorouracil/cisplatin-based chemoradiotherapy in Japanese patients with esophageal squamous cell carcinoma. Int J Med Sci 2012; 9: 833–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Yang PW, Hsieh MS, Huang YC, et al. Genetic variants of EGF and VEGF predict prognosis of patients with advanced esophageal squamous cell carcinoma. PLoS One 2014; 9: e100326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Lorenzen S, Panzram B, Keller G, et al. Association of the VEGF 936C>T polymorphism with FDG uptake, clinical, histopathological, and metabolic response in patients with adenocarcinomas of the esophagogastric junction. Mol Imaging Biol 2011; 13: 178–186. [DOI] [PubMed] [Google Scholar]
  • 115.Bradbury PA, Zhai R, Ma C, et al. Vascular endothelial growth factor polymorphisms and esophageal cancer prognosis. Clin Cancer Res 2009; 15: 4680–4685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Motoyama S, Miura M, Hinai Y, et al. Interleukin-2–330T>G genetic polymorphism associates with prognosis following surgery for thoracic esophageal squamous cell cancer. Ann Surg Oncol 2011; 18: 1995–2002. [DOI] [PubMed] [Google Scholar]
  • 117.Tang RG, Huang YZ, Yao LM, et al. Polymorphisms of transforming growth factor beta 1 (RS#1800468 and RS#1800471) and esophageal squamous cell carcinoma among Zhuangese population, China. Gene 2013; 512: 1–5. [DOI] [PubMed] [Google Scholar]
  • 118.Wu C, Li D, Jia W, et al. Genome-wide association study identifies common variants in SLC39A6 associated with length of survival in esophageal squamous-cell carcinoma. Nat Genet 2013; 45: 632–638. [DOI] [PubMed] [Google Scholar]
  • 119.Zhang R, Wang R, Zhang F, et al. Single nucleotide polymorphisms in the mitochondrial displacement loop and outcome of esophageal squamous cell carcinoma. J Exp Clin Cancer Res 2010; 29: 155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Umar M, Upadhyay R, Kumar S, et al. CASP8–652 6N del and CASP8 IVS12–19G>A gene polymorphisms and susceptibility/prognosis of ESCC: a case control study in northern Indian population. J Surg Oncol 2011; 103: 716–723. [DOI] [PubMed] [Google Scholar]
  • 121.Lee JM, Wu MT, Lee YC, et al. Association of GSTP1 polymorphism and survival for esophageal cancer. Clin Cancer Res 2005; 11: 4749–4753. [DOI] [PubMed] [Google Scholar]
  • 122.Wu X, Gu J, Wu TT, et al. Genetic variations in radiation and chemotherapy drug action pathways predict clinical outcomes in esophageal cancer. J Clin Oncol 2006; 24: 3789–3798. [DOI] [PubMed] [Google Scholar]
  • 123.Lee JM, Yang SY, Yang PW, et al. Polymorphism in epidermal growth factor receptor intron 1 predicts prognosis of patients with esophageal cancer after chemoradiation and surgery. Ann Surg Oncol 2011; 18: 2066–2073. [DOI] [PubMed] [Google Scholar]
  • 124.Alakus H, Bollschweiler E, Holscher AH, et al. Homozygous GNAS 393C-allele carriers with locally advanced esophageal cancer fail to benefit from platinum-based preoperative chemoradiotherapy. Ann Surg Oncol, 2014. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 125.Vashist YK, Kutup A, Musici S, et al. The GNAS1 T393C single nucleotide polymorphism predicts the natural postoperative course of complete resected esophageal cancer. Cell Oncol (Dordr) 2011; 34: 281–288. [DOI] [PubMed] [Google Scholar]
  • 126.Alakus H, Warnecke-Eberz U, Bollschweiler E, et al. GNAS1 T393C polymorphism is associated with histopathological response to neoadjuvant radiochemotherapy in esophageal cancer. Pharmacogenomics J 2009; 9: 202–207. [DOI] [PubMed] [Google Scholar]
  • 127.Kaifi JT, Rawnaq T, Schurr PG, et al. Short tandem repeat polymorphism in exon 4 of esophageal cancer-related gene 2 detected in genomic DNA is a prognostic marker for esophageal cancer. Am J Surg 2007; 194: 380–384. [DOI] [PubMed] [Google Scholar]
  • 128.Lu C, Xie H, Wang F, et al. Diet folate, DNA methylation and genetic polymorphisms of MTHFR C677T in association with the prognosis of esophageal squamous cell carcinoma. BMC Cancer 2011; 11: 91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Umar M, Upadhyay R, Khurana R, et al. Evaluation of MTHFR677C>T polymorphism in prediction and prognosis of esophageal squamous cell carcinoma: a case-control study in a northern Indian population. Nutr Cancer 2010; 62: 743–749. [DOI] [PubMed] [Google Scholar]
  • 130.Narumiya K, Metzger R, Bollschweiler E, et al. Impact of ABCB1 C3435T polymorphism on lymph node regression in multimodality treatment of locally advanced esophageal cancer. Pharmacogenomics 2011; 12: 205–214. [DOI] [PubMed] [Google Scholar]
  • 131.Lurje G, Leers JM, Pohl A, et al. Genetic variations in angiogenesis pathway genes predict tumor recurrence in localized adenocarcinoma of the esophagus. Ann Surg 2010; 251: 857–864. [DOI] [PubMed] [Google Scholar]
  • 132.Shibagaki I, Shimada Y, Wagata T, et al. Allelotype analysis of esophageal squamous cell carcinoma. Cancer Res 1994; 54: 2996–3000. [PubMed] [Google Scholar]
  • 133.Hildebrandt MA, Yang H, Hung MC, et al. Genetic variations in the PI3K/PTEN/AKT/mTOR pathway are associated with clinical outcomes in esophageal cancer patients treated with chemoradiotherapy. J Clin Oncol 2009; 27: 857–871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Azim K, McManus R, Brophy K, et al. Genetic polymorphisms and the risk of infection following esophagectomy. Positive association with TNF-alpha gene -308 genotype. Ann Surg 2007; 246: 122–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Lee JM, Lo AC, Yang SY, et al. Association of angiotensin-converting enzyme insertion/deletion polymorphism with serum level and development of pulmonary complications following esophagectomy. Ann Surg 2005; 241: 659–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Motoyama S, Miura M, Hinai Y, et al. Interferon-gamma 874A>T genetic polymorphism is associated with infectious complications following surgery in patients with thoracic esophageal cancer. Surgery 2009; 146: 931–938. [DOI] [PubMed] [Google Scholar]
  • 137.Metzger R, Warnecke-Eberz U, Alakus H, et al. Neoadjuvant radiochemotherapy in adenocarcinoma of the esophagus: ERCC1 gene polymorphisms for prediction of response and prognosis. J Gastrointest Surg 2012; 16: 26–34; discussion 34. [DOI] [PubMed] [Google Scholar]
  • 138.Hattori K, Kajiyama Y, Tsurumaru M. Mutation of the p53 gene predicts lymph node metastases in Japanese patients with esophageal carcinoma: DNA and immunohistochemical analyses. Dis Esophagus 2003; 16: 301–306. [DOI] [PubMed] [Google Scholar]
  • 139.Wang XC, Wu YP, Ye B, et al. Suppression of anoikis by SKP2 amplification and overexpression promotes metastasis of esophageal squamous cell carcinoma. Mol Cancer Res 2009; 7: 12–22. [DOI] [PubMed] [Google Scholar]
  • 140.Yang YL, Chu JY, Luo ML, et al. Amplification of PRKCI, located in 3q26, is associated with lymph node metastasis in esophageal squamous cell carcinoma. Genes Chromosomes Cancer 2008; 47: 127–136. [DOI] [PubMed] [Google Scholar]
  • 141.Reichelt U, Duesedau P, Tsourlakis M, et al. Frequent homogeneous HER-2 amplification in primary and metastatic adenocarcinoma of the esophagus. Mod Pathol 2007; 20: 120–129. [DOI] [PubMed] [Google Scholar]
  • 142.al-Kasspooles M, Moore JH, Orringer MB, et al. Amplification and over-expression of the EGFR and erbB-2 genes in human esophageal adenocarcinomas. Int J Cancer 1993; 54: 213–219. [DOI] [PubMed] [Google Scholar]
  • 143.Yang YL, Xu KL, Zhou Y, et al. Correlation of epidermal growth factor receptor overexpression with increased epidermal growth factor receptor gene copy number in esophageal squamous cell carcinomas. Chin Med J (Engl) 2012; 125: 450–454. [PubMed] [Google Scholar]
  • 144.Qin YR, Fu L, Sham PC, et al. Single-nucleotide polymorphism-mass array reveals commonly deleted regions at 3p22 and 3p14.2 associate with poor clinical outcome in esophageal squamous cell carcinoma. Int J Cancer 2008; 123: 826–830. [DOI] [PubMed] [Google Scholar]
  • 145.Huang XP, Wei F, Liu XY, et al. Allelic loss on 13q in esophageal squamous cell carcinomas from northern China. Cancer Lett 2002; 185: 87–94. [DOI] [PubMed] [Google Scholar]
  • 146.Harada H, Tanaka H, Shimada Y, et al. Lymph node metastasis is associated with allelic loss on chromosome 13q12–13 in esophageal squamous cell carcinoma. Cancer Res 1999; 59: 3724–3729. [PubMed] [Google Scholar]
  • 147.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. [DOI] [PubMed] [Google Scholar]
  • 148.Matsumoto Y, Nagasaka T, Kambara T, et al. Microsatellite instability and clinicopathological features in esophageal squamous cell cancer. Oncol Rep 2007; 18: 1123–1127. [PubMed] [Google Scholar]
  • 149.Wang K, Li J, Guo H, et al. MiR-196a binding-site SNP regulates RAP1A expression contributing to esophageal squamous cell carcinoma risk and metastasis. Carcinogenesis 2012; 33: 2147–2154. [DOI] [PubMed] [Google Scholar]
  • 150.Umar M, Upadhyay R, Kumar S, et al. Modification of risk, but not survival of esophageal cancer patients by esophageal cancer-related gene 1 Arg290Gln polymorphism: a case-control study and meta-analysis. J Gastroenterol Hepatol 2013; 28: 1717–1724. [DOI] [PubMed] [Google Scholar]
  • 151.Miao X, Sun T, Wang Y, et al. Functional STK15 Phe31Ile polymorphism is associated with the occurrence and advanced disease status of esophageal squamous cell carcinoma. Cancer Res 2004; 64: 2680–2683. [DOI] [PubMed] [Google Scholar]
  • 152.Begg CB, Mazumdar M. Operating characteristics of a rank correlation test for publication bias. Biometrics 1994; 50: 1088–1101. [PubMed] [Google Scholar]
  • 153.Thompson SG, Higgins JP. How should meta-regression analyses be undertaken and interpreted? Stat Med 2002; 21: 1559–1573. [DOI] [PubMed] [Google Scholar]
  • 154.Talsma K, van Hagen P, Grotenhuis BA, et al. Comparison of the 6th and 7th Editions of the UICC-AJCC TNM Classification for Esophageal Cancer. Ann Surg Oncol 2012; 19: 2142–2148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Olivier M, Langerod A, Carrieri P, et al. The clinical value of somatic TP53 gene mutations in 1,794 patients with breast cancer. Clin Cancer Res 2006; 12: 1157–1167. [DOI] [PubMed] [Google Scholar]
  • 156.Smith FM, Stephens RB, Kennedy MJ, et al. P53 abnormalities and outcomes in colorectal cancer: a systematic review. Br J Cancer 2005; 92: 1813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Xu J, Reumers J, Couceiro JR, et al. Gain of function of mutant p53 by coaggregation with multiple tumor suppressors. Nat Chem Biol 2011; 7: 285–295. [DOI] [PubMed] [Google Scholar]
  • 158.Lehmann S, Bykov VJ, Ali D, et al. Targeting p53 in vivo: a first-in-human study with p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer. J Clin Oncol 2012; 30: 3633–3639. [DOI] [PubMed] [Google Scholar]
  • 159.Bellini MF, Cadamuro AC, Succi M, et al. Alterations of the TP53 gene in gastric and esophageal carcinogenesis. J Biomed Biotechnol 2012; 2012: 891961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Lai PB, Chi TY, Chen GG. Different levels of p53 induced either apoptosis or cell cycle arrest in a doxycycline-regulated hepatocellular carcinoma cell line in vitro. Apoptosis 2007; 12: 387–393. [DOI] [PubMed] [Google Scholar]
  • 161.Coffill CR, Muller PA, Oh HK, et al. Mutant p53 interactome identifies nardilysin as a p53R273H-specific binding partner that promotes invasion. EMBO Rep 2012; 13: 638–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Chen M, Huang J, Zhu Z, et al. Systematic review and meta-analysis of tumor biomarkers in predicting prognosis in esophageal cancer. BMC Cancer 2013; 13: 539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Zhang SS, Huang QY, Yang H, et al. Correlation of p53 status with the response to chemotherapy-based treatment in esophageal cancer: a meta-analysis. Ann Surg Oncol 2013; 20: 2419–2427. [DOI] [PubMed] [Google Scholar]
  • 164.Eguchi K, Yao T, Konomoto T, et al. Discordance of p53 mutations of synchronous colorectal carcinomas. Mod Pathol 2000; 13: 131–139. [DOI] [PubMed] [Google Scholar]
  • 165.Sim NL, Kumar P, Hu J, et al. SIFT web server: predicting effects of amino acid substitutions on proteins. Nucleic Acids Res 2012; 40: W452–W457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Adzhubei IA, Schmidt S, Peshkin L, et al. A method and server for predicting damaging missense mutations. Nat Methods 2010; 7: 248–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010; 376: 687–697. [DOI] [PubMed] [Google Scholar]
  • 168.Bible KC, Peethambaram PP, Oberg AL, et al. A phase 2 trial of flavopiridol (Alvocidib) and cisplatin in platin-resistant ovarian and primary peritoneal carcinoma: MC0261. Gynecol Oncol 2012; 127: 55–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Luke JJ, D'Adamo DR, Dickson MA, et al. The cyclin-dependent kinase inhibitor flavopiridol potentiates doxorubicin efficacy in advanced sarcomas: preclinical investigations and results of a phase I dose-escalation clinical trial. Clin Cancer Res 2012; 18: 2638–2647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Lorenzen S, Schuster T, Porschen R, et al. Cetuximab plus cisplatin-5-fluorouracil versus cisplatin-5-fluorouracil alone in first-line metastatic squamous cell carcinoma of the esophagus: a randomized phase II study of the Arbeitsgemeinschaft Internistische Onkologie. Ann Oncol 2009; 20: 1667–1673. [DOI] [PubMed] [Google Scholar]
  • 171.Rodriguez CP, Adelstein DJ, Rice TW, et al. A phase II study of perioperative concurrent chemotherapy, gefitinib, and hyperfractionated radiation followed by maintenance gefitinib in locoregionally advanced esophagus and gastroesophageal junction cancer. J Thorac Oncol 2010; 5: 229–235. [DOI] [PubMed] [Google Scholar]
  • 172.Idelevich E, Kashtan H, Klein Y, et al. Prospective phase II study of neoadjuvant therapy with cisplatin, 5-fluorouracil, and bevacizumab for locally advanced resectable esophageal cancer. Onkologie 2012; 35: 427–431. [DOI] [PubMed] [Google Scholar]
  • 173.De Vita F, Orditura M, Martinelli E, et al. A multicenter phase II study of induction chemotherapy with FOLFOX-4 and cetuximab followed by radiation and cetuximab in locally advanced oesophageal cancer. Br J Cancer 2011; 104: 427–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Walther A, Houlston R, Tomlinson I. Association between chromosomal instability and prognosis in colorectal cancer: a meta-analysis. Gut 2008; 57: 941–950. [DOI] [PubMed] [Google Scholar]
  • 175.Zhou W, Gurubhagavatula S, Liu G, et al. Excision repair cross-complementation group 1 polymorphism predicts overall survival in advanced non-small cell lung cancer patients treated with platinum-based chemotherapy. Clin Cancer Res 2004; 10: 4939–4943. [DOI] [PubMed] [Google Scholar]
  • 176.Yin M, Yan J, Martinez-Balibrea E, et al. ERCC1 and ERCC2 polymorphisms predict clinical outcomes of oxaliplatin-based chemotherapies in gastric and colorectal cancer: a systemic review and meta-analysis. Clin Cancer Res 2011; 17: 1632–1640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Giovannetti E, Pacetti P, Reni M, et al. Association between DNA-repair polymorphisms and survival in pancreatic cancer patients treated with combination chemotherapy. Pharmacogenomics 2011; 12: 1641–1652. [DOI] [PubMed] [Google Scholar]
  • 178.Li Y, Liu Z, Liu H, et al. ERCC1 and ERCC2 variants predict survival in gastric cancer patients. PLoS One 2013; 8: e71994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Hoffmeyer S, Burk O, von Richter O, et al. Functional polymorphisms of the human multidrug-resistance gene: multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc Natl Acad Sci USA 2000; 97: 3473–3478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Wu H, Kang H, Liu Y, et al. Association of ABCB1 genetic polymorphisms with susceptibility to colorectal cancer and therapeutic prognosis. Pharmacogenomics 2013; 14: 897–911. [DOI] [PubMed] [Google Scholar]
  • 181.Bond GL, Hu W, Bond EE, et al. A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell 2004; 119: 591–602. [DOI] [PubMed] [Google Scholar]
  • 182.Ma Y, Bian J, Cao H. MDM2 SNP309 rs2279744 polymorphism and gastric cancer risk: a meta-analysis. PLoS One 2013; 8: e56918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Horie N, Takeishi K. Functional structure of the promoter region of the human thymidylate synthase gene and nuclear factors that regulate the expression of the gene. Nucleic Acids Symp Ser 1995; 0: 77–78. [PubMed] [Google Scholar]
  • 184.Lima A, Seabra V, Martins S, et al. Thymidylate synthase polymorphisms are associated to therapeutic outcome of advanced non-small cell lung cancer patients treated with platinum-based chemotherapy. Mol Biol Rep 2014; 41: 3349–3357. [DOI] [PubMed] [Google Scholar]
  • 185.Frosst P, Blom HJ, Milos R, et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet 1995; 10: 111–113. [DOI] [PubMed] [Google Scholar]
  • 186.Boccia S, Hung R, Ricciardi G, et al. Meta- and pooled analyses of the methylenetetrahydrofolate reductase C677T and A1298C polymorphisms and gastric cancer risk: a huge-GSEC review. Am J Epidemiol 2008; 167: 505–516. [DOI] [PubMed] [Google Scholar]
  • 187.Abdel-Rahman SZ, El-Zein RA. The 399Gln polymorphism in the DNA repair gene XRCC1 modulates the genotoxic response induced in human lymphocytes by the tobacco-specific nitrosamine NNK. Cancer Lett 2000; 159: 63–71. [DOI] [PubMed] [Google Scholar]
  • 188.Cui Z, Yin Z, Li X, et al. Association between polymorphisms in XRCC1 gene and clinical outcomes of patients with lung cancer: a meta-analysis. BMC Cancer 2012; 12: 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Yu JJ, Lee KB, Mu C, et al. Comparison of two human ovarian carcinoma cell lines (A2780/CP70 and MCAS) that are equally resistant to platinum, but differ at codon 118 of the ERCC1 gene. Int J Oncol 2000; 16: 555–560. [DOI] [PubMed] [Google Scholar]
  • 190.Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 2012; 366: 883–892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Baylin SB, Ohm JE. Epigenetic gene silencing in cancer —a mechanism for early oncogenic pathway addiction? Nat Rev Cancer 2006; 6: 107–116. [DOI] [PubMed] [Google Scholar]
  • 192.Simon R, Roychowdhury S. Implementing personalized cancer genomics in clinical trials. Nat Rev Drug Discov 2013; 12: 358–369. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Data

Articles from Annals of Oncology are provided here courtesy of Oxford University Press

RESOURCES