A more detailed understanding of the somatic genetic events that drive gastrointestinal adenocarcinomas is necessary to improve diagnosis and therapy. Using data from high-density genomic profiling arrays, we conducted an analysis of somatic copy-number aberrations in 486 gastrointestinal adenocarcinomas including 296 esophageal and gastric cancers. Focal amplifications were substantially more prevalent in gastric/esophageal adenocarcinomas than colorectal tumors. We identified 64 regions of significant recurrent amplification and deletion, some shared and others unique to the adenocarcinoma types examined. Amplified genes were noted in 37% of gastric/esophageal tumors, including in therapeutically targetable kinases such as ERBB2, FGFR1, FGFR2, EGFR, and MET, suggesting the potential use of genomic amplifications as biomarkers to guide therapy of gastric and esophageal cancers where targeted therapeutics have been less developed compared with colorectal cancers. Amplified loci implicated genes with known involvement in carcinogenesis but also pointed to regions harboring potentially novel cancer genes, including a recurrent deletion found in 15% of esophageal tumors where the Runt transcription factor subunit RUNX1 was implicated, including by functional experiments in tissue culture. Together, our results defined genomic features that were common and distinct to various gut-derived adenocarcinomas, potentially informing novel opportunities for targeted therapeutic interventions. Cancer Res; 72(17); 4383–93. ©2012 AACR.

Colorectal, gastric, and esophageal adenocarcinomas collectively account for approximately 180,000 cancer diagnoses and 76,500 deaths each year in the United States and approximately 1.3 million deaths worldwide (1, 2). A better understanding of the somatic genetics of these diseases is a prerequisite for earlier diagnosis and more effective treatment. Colorectal cancer (CRC) genomes have been studied extensively (3, 4); the value of this information is realized by persuasive evidence that KRAS and BRAF mutations in CRC predict lack of response to cetuximab (5, 6). Gastric cancer (GC) and especially esophageal adenocarcinoma (EA) has been subjected to fewer large-scale studies (7–9).

Cancers of the esophagus and stomach commonly arise in a background of intestinal metaplasia, but develop within distinct luminal environments. Nevertheless, they often are treated with identical chemotherapy, and many clinical trials combine patients with these 2 diseases (10, 11). Although the process of intestinal metaplasia preceding GC and EA suggests that these tumors may resemble adenocarcinomas arising from the intestine, they show distinct clinical behavior from CRC. It is therefore important to define the similarities and differences among digestive tract adenocarcinomas at the genomic and molecular levels. Such a comparison can inform both mechanistic studies and strategies for biomarker-driven therapy.

Two challenges exist in the somatic genetic analysis of cancer: (i) distinguishing “driver” alterations that contribute to tumor development, maintenance, or proliferation from random “passenger” alterations that do not contribute to the neoplastic process, and (ii) identifying the specific genes that mediate tumor progression. Both challenges must be confronted in analysis of somatic copy-number alterations (SCNA) as tumors often harbor many such alterations, each of which often encompasses up to thousands of genes. The study of SCNAs has been greatly enhanced by high-density genomic arrays allowing resolution of individual SCNA boundaries and the ability to study large numbers of tumors. Statistical analysis of SCNAs across many samples can identify regions altered more frequently than expected by chance and also pinpoint the most likely culprit genes in these regions. Pooling data from different but related cancer types can increase both statistical power and the ability to resolve specific gene targets of SCNAs. Given the related origins and documented shared copy-number characteristics of gut-derived adenocarcinomas (12), we hypothesized that evaluating genomic events across these tumors will increase our power to identify common genes active in gut adenocarcinomas and also help uncover differences. Here, we report the largest analysis of SCNAs across gut adenocarcinoma genomes and systematically compare significantly recurrent structural genetic alterations in tumors from distinct regions of the gut. We find multiple known and novel recurrent alterations, including region-specific and shared events.

All samples were fresh frozen primary resections from patients not treated with prior chemotherapy or radiation. All cases had diagnoses confirmed by pathologic review and only cases with estimated carcinoma content >70% were selected (Supplementary Table S1). The sample set was not enriched for other features. Tumors annotated as having originated from the gastric–esophageal junction were assigned to the EA collection. DNA was extracted (Supplementary Table S1), quantified with Picogreen dye and hybridized to (215 samples) GeneChip Human Mapping StyI 250K arrays (Affymetrix) or (271 samples) Genome-Wide Human SNP Array 6.0 (SNP6.0; Affymetrix) genomic profiling arrays, according to the manufacturer's instructions. Data from each of the 2 array platforms were independently normalized and segmented using all data present upon each of the 2 platforms (12, 13). Regions of known germline copy-number polymorphisms were then removed as previously described (14). Human genome build hg18 was used, and raw data files have been deposited at the Gene Expression Omnibus (GSE36460).

Significantly recurrent SCNAs were identified using GISTIC 2.0. (15) All data from each array were used to generate SCNA profiles for each tumor. To enable probe bound GISTIC analyses across data from 2 array platforms, the segmented data from each sample was remapped to the 196,800 probes shared by the 2 platforms. In some cases, this remapping modified the position of the probe bounding the transition between 2 copy-number segments. In these cases, the boundaries were remapped to the nearest probes in the joint set. To remove potentially spurious SCNAs, segments defined by fewer than 9 shared probes were removed (16). Additional details are described in Supplementary Methods.

A comprehensive copy-number dataset from digestive tract adenocarcinomas

We analyzed a cohort of 363 new and 123 publically available genome array profiles from primary untreated gut adenocarcinomas including EA (186), GC (110), and CRC (190; Supplementary Table S1). We determined genome-wide copy-number profiles using either 250K StyI (238,000 probes) or SNP6.0 genome arrays (1.8 million probes; Supplementary Table S1). Copy-number alterations were identified using the full complement of data from each array type. To enable the analysis across platforms, the segmented copy-number data from each sample were remapped to the 196,800 probes common to both arrays. We found no evidence of bias introduced by pooling samples from the 2 platforms (Supplementary Note S1).

Chromosomal instability increases from lower to upper gut adenocarcinomas

Across this set of adenocarcinoma SCNA profiles, visual inspection of the segmented data showed alterations in nearly every part of the genome, and variations in the amount of genomic disruption between different cancers and cancer types (Fig. 1A). To compare levels of genomic disruption, we separately evaluated the frequencies of arm-level (comprising half or more of a chromosome arm) and focal SCNAs in each cancer type. Focal alterations were noted to occur throughout the chromosomes, but showed some predilection for the regions closer to the centromeres and telomeres of each chromosome (Supplementary Fig. S1).

Figure 1.

Patterns of chromosomal instability differ across adenocarcinomas of the gastrointestinal tract. A, amplifications (red) and deletions (blue) are displayed for 486 gut adenocarcinomas (x-axis) across the genome (y-axis). B and C, quantitation of arm-level (B) and focal (C) SCNAs. Solid bars represent median numbers of events per sample. D, quantitation of inferred multicopy SCNAs (high-level amplifications with inferred copy >4 and presumable homozygous deletions, inferred copy-number <1.3). ***, P < 0.0005; **, P < 0.005; *, P < 0.05; n.s., not significant.

Figure 1.

Patterns of chromosomal instability differ across adenocarcinomas of the gastrointestinal tract. A, amplifications (red) and deletions (blue) are displayed for 486 gut adenocarcinomas (x-axis) across the genome (y-axis). B and C, quantitation of arm-level (B) and focal (C) SCNAs. Solid bars represent median numbers of events per sample. D, quantitation of inferred multicopy SCNAs (high-level amplifications with inferred copy >4 and presumable homozygous deletions, inferred copy-number <1.3). ***, P < 0.0005; **, P < 0.005; *, P < 0.05; n.s., not significant.

Close modal

The median number of arm-level gains varied little between types (Fig. 1B), but was significantly increased in focal amplifications in EA and GC compared with CRC (Fig. 1C). Some tumors, particularly among CRC and GC types, showed little apparent genomic disruption, potentially attributable to microsatellite instability or stromal contamination (Supplementary Note S2). The enhanced rate of focal amplification in EA and GC compared with CRC remained valid when these genomically quiet tumors were removed from analysis (Supplementary Fig. S2). Upper gut adenocarcinomas exhibited an even greater excess of higher-level, multicopy focal amplifications (Fig. 1D), which remained after we accounted for possible contaminating noncancer DNA through use of sample-specific thresholds for defining these events (Supplementary Fig. S3). The higher rates of focal genomic amplifications in EA and GC relative to CRC suggest that underlying mechanisms of genomic instability may differ between upper and lower gastrointestinal adenocarcinomas and that genomic amplification may be a more common means of oncogene activation in GC/EA.

The difference in rates of focal amplifications between upper and lower gastrointestinal adenocarcinomas did not hold for deletions. Fewer arm-level deletions were seen in GC than other gut adenocarcinomas (Fig. 1B). There was a modest increase in focal deletions in EA compared with GC and CRC (Fig. 1B), a trend that persisted when cases without arm-level SCNAs were excluded (Supplementary Fig. S2). EA and CRC exhibit similar rates of multicopy deletions that may represent homozygous deletions, rates that are significantly higher than in GC (Fig. 1D). This pattern persisted after exclusion of “quiet” samples and use of sample-specific thresholds to identify the multicopy deletions (Supplementary Fig. S3). The discordant bias toward focal amplifications but not deletions in upper gastrointestinal cancers suggests that the mechanisms and selective pressures leading to amplification may differ from deletion.

Recurrent amplifications and deletions across the pooled dataset

Arm-level events.

We next carried out a GISTIC 2.0 analysis to define significantly recurrent SCNAs, starting with arm-level SCNAs. Arm-level amplifications of chromosomes 7p, 8q, 20p, and 20q recurred significantly across all 3 cancer types (Supplementary Fig. S4A). Events restricted to specific subtypes included 1q gains in GC and 13q gains in CRC and GC. These significant arm-level gains have been observed previously (17–23). Arm-level deletions were more variable across tumor types. In GC, deletions of 4p and 4q alone were significant reflecting a lower degree of arm-level losses detected earlier in GC (Supplementary Fig. S4B), but these and other deletions (8p, 18p, and 18q) were also detected in EA and CRC. Loss of arms 8p, 14q, and 15q were of higher significance in CRC, whereas loss of 5q, 9p, and 21q were particularly significant among EAs. Loss of 17p (containing TP53) was significant in EA and CRC, but not GC. Unique significant losses of 9p and 21q in EA are notable because these arms respectively contain the known and putative tumor suppressors CDKN2A and RUNX1, both targets of focal deletion in EA.

These results are in accordance with previously published data. We compared the frequency of alteration for each cytoband in each cancer type to frequencies determined across 998 CRC, 741 GC, and 71 EA in the Progenetix database curated from cCGH and aCGH data (24, 25). The cytoband-level data are similar for all cancer types, with the exception of lower frequencies of 17q gain in our GC and EA samples (Supplementary Fig. S5). Notably, the low frequency of deletions in GC data compiled by Progenetix mirrored our data suggesting that result is not unique to the samples in our collection.

Focal alterations.

We next evaluated focal SCNAs across all 486 tumors and identified 33 regions subject to significant (q < 0.01) focal amplification (Fig. 2 and Table 1). Thirteen of these regions contain known oncogenes, including 4 genes involved in cell-cycle regulation (CCNE1, CCND1, CDK6, and MYC) and 7 members of tyrosine kinase/MAPK signaling pathways (EGFR, KRAS, MET, ERBB2, FGFR1, FGFR2, and IGF1R). Twenty significant peaks contained no established oncogenes, suggesting potential presence of novel genes or noncoding transcripts that promote intestinal metaplasia and/or gastrointestinal carcinogenesis.

Figure 2.

Identification of significant focal SCNAs in a cohort of esophageal, gastric, and colorectal adenocarcinomas. GISTIC q-values (x-axis) for deletions (left, blue) and amplifications (right, red) are plotted across the genome (y-axis). The 10 most significant peaks are labeled with known or putative gene targets. Values in parentheses represent the number of genes in the peak region. *, genes immediately adjacent to the peak.

Figure 2.

Identification of significant focal SCNAs in a cohort of esophageal, gastric, and colorectal adenocarcinomas. GISTIC q-values (x-axis) for deletions (left, blue) and amplifications (right, red) are plotted across the genome (y-axis). The 10 most significant peaks are labeled with known or putative gene targets. Values in parentheses represent the number of genes in the peak region. *, genes immediately adjacent to the peak.

Close modal
Table 1.

Significantly amplified focal SCNAs in a cohort of esophageal, gastric, and colorectal adenocarcinomas (q-value <0.01)

Significant amplifications (across all gut adenocarcinomas)
CytobandResidual q-valuePeak boundaries, MbNumber of genes in peakCandidate target(s)Gut adenocarcinoma types represented
Gut adenocarcinomas 
 12p12.1 8.04E−51 25.23–25.34 KRAS E, G 
 18q11.2 5.05E−37 17.95–18.05 GATA6 E, G, C 
 17q12 4.34E−35 34.97–35.27 10 ERBB2 E, G, C 
 19q12 5.97E−34 34.95–35.10 CCNE1 E, G 
 8q24.21 8.23E−34 128.50–128.83 MYC E, G, C 
 8p23.1 2.10E−32 11.41–11.71 GATA4 E, G 
 11q13.2 5.26E−22 68.97–69.49 CCND1, FGF3, FGF4, FGF19 E, G 
 7q21.2 3.36E−17 92.32–92.50 CDK6* 
 6p21.1 2.12E−16 43.79–43.99 VEGFA* E, G 
 7p11.2 6.71E−16 54.92–55.28 EGFR E, G 
 17q21.2 2.23E−11 37.02–37.21  
 9p13.3 7.03E−09 35.52–35.93 24  
 12q15 6.23E−08 67.09–68.25 10 MDM2, FRS2 E, G 
 7q22.1 1.41E−07 98.41–99.02 16  
 13q13.1 2.04E−06 32.18–33.33  
 10q22.2 1.10E−05 75.00–75.80 16  
 7q31.2 1.32E−05 115.98–116.42 MET 
 1q21.3 1.69E−05 146.23–150.89 106 MCL1 
 1q42.3 1.71E−04 233.02–233.42  
 10q26.12 3.17E−04 122.75–123.37 FGFR2 
 13q14.11 3.67E−04 40.27–40.83 10  
 13q22.1 3.90E−04 72.47–73.00 KLF5 None 
 6q23.3 8.14E−04 135.32–135.87 MYB 
 7q34 1.03E−03 141.92–142.26 EPHB6 
 8p12 1.28E−03 38.23–38.46 FGFR1 
 3q26.2 1.55E−03 169.95–175.23 32 PRKCι 
 11p14.2 1.81E−03 26.94–27.57  
 1p36.22 1.99E−03 103.10–110.18 10  
 15q26.1 3.52E−03 86.15–100.34 94 IGF1R None 
 17q24.3 4.18E−03 67.82–68.05 SOX9 None 
 11q14.1 4.19E−03 75.12–79.48 31 GAB2 None 
 20q13.2 6.89E−03 51.55–52.11  None 
 3q27.1 8.69E−03 185.72–186.17 EPHB3 
Significant amplifications (across all gut adenocarcinomas)
CytobandResidual q-valuePeak boundaries, MbNumber of genes in peakCandidate target(s)Gut adenocarcinoma types represented
Gut adenocarcinomas 
 12p12.1 8.04E−51 25.23–25.34 KRAS E, G 
 18q11.2 5.05E−37 17.95–18.05 GATA6 E, G, C 
 17q12 4.34E−35 34.97–35.27 10 ERBB2 E, G, C 
 19q12 5.97E−34 34.95–35.10 CCNE1 E, G 
 8q24.21 8.23E−34 128.50–128.83 MYC E, G, C 
 8p23.1 2.10E−32 11.41–11.71 GATA4 E, G 
 11q13.2 5.26E−22 68.97–69.49 CCND1, FGF3, FGF4, FGF19 E, G 
 7q21.2 3.36E−17 92.32–92.50 CDK6* 
 6p21.1 2.12E−16 43.79–43.99 VEGFA* E, G 
 7p11.2 6.71E−16 54.92–55.28 EGFR E, G 
 17q21.2 2.23E−11 37.02–37.21  
 9p13.3 7.03E−09 35.52–35.93 24  
 12q15 6.23E−08 67.09–68.25 10 MDM2, FRS2 E, G 
 7q22.1 1.41E−07 98.41–99.02 16  
 13q13.1 2.04E−06 32.18–33.33  
 10q22.2 1.10E−05 75.00–75.80 16  
 7q31.2 1.32E−05 115.98–116.42 MET 
 1q21.3 1.69E−05 146.23–150.89 106 MCL1 
 1q42.3 1.71E−04 233.02–233.42  
 10q26.12 3.17E−04 122.75–123.37 FGFR2 
 13q14.11 3.67E−04 40.27–40.83 10  
 13q22.1 3.90E−04 72.47–73.00 KLF5 None 
 6q23.3 8.14E−04 135.32–135.87 MYB 
 7q34 1.03E−03 141.92–142.26 EPHB6 
 8p12 1.28E−03 38.23–38.46 FGFR1 
 3q26.2 1.55E−03 169.95–175.23 32 PRKCι 
 11p14.2 1.81E−03 26.94–27.57  
 1p36.22 1.99E−03 103.10–110.18 10  
 15q26.1 3.52E−03 86.15–100.34 94 IGF1R None 
 17q24.3 4.18E−03 67.82–68.05 SOX9 None 
 11q14.1 4.19E−03 75.12–79.48 31 GAB2 None 
 20q13.2 6.89E−03 51.55–52.11  None 
 3q27.1 8.69E−03 185.72–186.17 EPHB3 

NOTE: Bold designates therapeutic target.

*, genes immediately adjacent to the peak.

The 18q11.2 amplification peak is the second most significant peak after KRAS and contains only the endodermal transcription factor GATA6. Coupled with functional data suggesting a role for GATA6 in esophageal carcinogenesis (26, 27), these results implicate GATA6 as an important contributor to gastrointestinal neoplasia. The sixth most significant peak in the composite dataset, located at 8p23.1, contains 4 genes, including the related transcription factor GATA4, a candidate target noted previously (28).

We also identified 30 regions of significant focal deletion (Fig. 2 and Table 2). Eight of these regions include genes such as FHIT and WWOX, with exons spread over large genomic loci (in excess of 1 Mb). Prior studies suggest that such regions often lie in “fragile sites” or areas of low gene density where deletion may be tolerated, and may not harbor functional tumor suppressors (12, 29). An additional 8 regions contain the known tumor suppressors CDKN2A, SMAD4, PTEN, APC, RUNX1, ARID1A, and ATM and the putative tumor suppressor PARD3B (30). Fourteen regions did not contain known tumor suppressors or large-footprint genes, but could contain novel factors whose loss contributes to intestinal metaplasia or cancer. Our analysis would not have detected regions of loss of heterozygosity that did not lead to copy-number loss.

Table 2.

Significantly deleted focal SCNAs in a cohort of esophageal, gastric, and colorectal adenocarcinomas (q-value <0.01)

Significant deletions (across all gut adenocarcinomas)
CytobandResidual q-valuePeak boundaries, MbNumber of genes in peakCandidate target(s)Gut adenocarcinoma types represented
Gut adenocarcinomas 
 3p14.2 1.207E−313 58.98–61.52 FHIT E, G, C 
 16q23.1 2.96E−151 76.69–78.21 WWOX E, G, C 
 9p21.3 5.08E−56 21.98–22.01 CDKN2A 
 20p12.1 3.22E−50 14.21–15.99 MACROD2 E, G, C 
 5q11.2 9.09E−38 58.30–59.84 PDE4D E, G, C 
 16p13.3 8.41E−36 50.63–77.09 A2BP1 
 6p25.3 2.09E−28 1.54–2.57  E, G 
 4q22.1 2.60E−27 91.37–93.49 FAM190A E, G, C 
 6q26 3.61E−21 161.61–163.13 PARK2 E, G, C 
 9p24.1 1.32E−14 7.77–12.71 PTPRD E, G 
 18q21.2 7.52E−14 46.77–46.97 SMAD4 E, C 
 1p36.11 1.36E−11 26.77–27.31 11 ARID1A E, C 
 21q11.2 1.61E−06 1.00–1.64 17  
 1q44 2.64E−06 24.39–24.48  
 8p23.3 2.64E−06 1.00–1.71  
 10q23.31 2.64E−06 89.44–90.07 PTEN 
 21q22.12 5.76E−06 35.03–35.35 RUNX1 
 4q35.1 1.80E−05 179.06–187.23 31  
 7q36.3 3.01E−05 156.49–158.82 12  
 5q22.2 5.22E−05 110.88–113.74 12 APC 
 1p13.2 8.53E−05 111.01–119.86 79  
 2q33.2 1.15E−04 204.53–206.27 PARD3B 
 3p26.2 1.24E−04 4.32–4.51  None 
 4q35.2 4.15E−04 185.37–191.27 36 CASP3 
 17p12 1.38E−03 11.40–12.51  None 
 11q22.3 2.26E−03 103.49–112.70 62 ATM 
 5q15 3.85E−03 83.69–106.74 56  None 
 21q22.3 8.17E−03 38.21–46.94 116  None 
 12q21.2 8.88E−03 71.30–86.91 39  None 
 10q11.23 9.24E−03 52.31–53.77  
Significant deletions (across all gut adenocarcinomas)
CytobandResidual q-valuePeak boundaries, MbNumber of genes in peakCandidate target(s)Gut adenocarcinoma types represented
Gut adenocarcinomas 
 3p14.2 1.207E−313 58.98–61.52 FHIT E, G, C 
 16q23.1 2.96E−151 76.69–78.21 WWOX E, G, C 
 9p21.3 5.08E−56 21.98–22.01 CDKN2A 
 20p12.1 3.22E−50 14.21–15.99 MACROD2 E, G, C 
 5q11.2 9.09E−38 58.30–59.84 PDE4D E, G, C 
 16p13.3 8.41E−36 50.63–77.09 A2BP1 
 6p25.3 2.09E−28 1.54–2.57  E, G 
 4q22.1 2.60E−27 91.37–93.49 FAM190A E, G, C 
 6q26 3.61E−21 161.61–163.13 PARK2 E, G, C 
 9p24.1 1.32E−14 7.77–12.71 PTPRD E, G 
 18q21.2 7.52E−14 46.77–46.97 SMAD4 E, C 
 1p36.11 1.36E−11 26.77–27.31 11 ARID1A E, C 
 21q11.2 1.61E−06 1.00–1.64 17  
 1q44 2.64E−06 24.39–24.48  
 8p23.3 2.64E−06 1.00–1.71  
 10q23.31 2.64E−06 89.44–90.07 PTEN 
 21q22.12 5.76E−06 35.03–35.35 RUNX1 
 4q35.1 1.80E−05 179.06–187.23 31  
 7q36.3 3.01E−05 156.49–158.82 12  
 5q22.2 5.22E−05 110.88–113.74 12 APC 
 1p13.2 8.53E−05 111.01–119.86 79  
 2q33.2 1.15E−04 204.53–206.27 PARD3B 
 3p26.2 1.24E−04 4.32–4.51  None 
 4q35.2 4.15E−04 185.37–191.27 36 CASP3 
 17p12 1.38E−03 11.40–12.51  None 
 11q22.3 2.26E−03 103.49–112.70 62 ATM 
 5q15 3.85E−03 83.69–106.74 56  None 
 21q22.3 8.17E−03 38.21–46.94 116  None 
 12q21.2 8.88E−03 71.30–86.91 39  None 
 10q11.23 9.24E−03 52.31–53.77  

The combined analysis across 3 tumor types enabled identification of less common SCNAs. Five amplification peaks were significant in the composite set but not in any individual cancer type. One of these, 13q22.1, contains only 2 genes, including the proliferative transcription factor KLF5. The combined dataset also enabled more precise identification of the likely targets of focal SCNAs. For example, the 1p36.11 deletion narrowed from 89 genes in the EA set to only 11 genes in the combined dataset, including the chromatin-modifying enzyme ARID1A, a recently identified target of frequent mutation in clear cell ovarian and gastric adenocarcinomas (31, 32). However, combining data across platforms also entailed some loss of resolution for the SNP6 data. We, therefore, carried out a separate analysis of the SNP6 data, which yielded similar results to the composite analysis, though with fewer peaks (Supplementary Note S3 and Table S6).

To identify relationships between genes targeted by focal alterations, we evaluated the co-occurrence or exclusivity of focal alterations at all GISTIC peak regions. After correcting for multiple hypothesis testing and tissue type, the only significant (Bonferroni-corrected P-value ≥0.05) findings were correlations of amplifications of CCNE1 with each of 2 peaks with unknown targets: a deletion peak at 6p25.3 and amplification of 1q42.3. These findings may suggest cooperativity between these novel events and amplification of CCNE1, or reflect subsets of tumors that for other reasons tend to share alterations in these regions.

Comparison of focal alterations across gut adenocarcinoma and other cancer types

Comparison of focal amplifications.

We analyzed focal alterations in each tumor type separately and identified 5, 14, and 25 amplification peaks in CRC, GC, and EA, respectively (Fig. 3A and Supplementary Fig. S6 and Table S2). Highlighting the similarities and differences among digestive tract cancers, only 3 amplifications were significant in all tumor types: 8q24.21 (containing MYC), 17q12 (containing ERBB2), and 18q11.2 (containing GATA6). Among these peaks, ERBB2 is amplified more commonly in esophageal (17%) and gastric (13%) than in colorectal (6%) tumors (Supplementary Fig. S7).

Figure 3.

Venn diagram of focal events common across gut adenocarcinomas. Significantly amplified (A) or deleted (B) peaks (or gene if likely candidate available) as determined by GISTIC analysis of focal alterations in each adenocarcinoma subtype. The color code denotes peaks that are unique to a given tumor type or shared between 2 or 3 of the adenocarcinoma subtypes. Bolded genes represent genes significantly altered and common across esophageal, gastric, and colorectal tumors. *, significant SCNAs in a dataset containing 3,131 cancers not including CRC, EA, or GC (12).

Figure 3.

Venn diagram of focal events common across gut adenocarcinomas. Significantly amplified (A) or deleted (B) peaks (or gene if likely candidate available) as determined by GISTIC analysis of focal alterations in each adenocarcinoma subtype. The color code denotes peaks that are unique to a given tumor type or shared between 2 or 3 of the adenocarcinoma subtypes. Bolded genes represent genes significantly altered and common across esophageal, gastric, and colorectal tumors. *, significant SCNAs in a dataset containing 3,131 cancers not including CRC, EA, or GC (12).

Close modal

Two amplification peaks were restricted to CRCs. One amplicon contains the RTK, FGFR1, not previously reported in this disease, but noted to be overexpressed (33). The other unique peak is adjacent to the CRC oncogene CDK8 at 13q12.2 (34). Esophageal and gastric adenocarcinomas shared 7 amplicons, containing VEGFA, EGFR, GATA4, CCND1, MDM2, CCNE1, and KRAS. The most significantly amplified gene across our dataset, KRAS, showed a strong foregut preference. Only 5% of CRCs carried focal KRAS amplification, compared with 21% of upper GI cancers. Conversely, CRCs have substantially higher rates of KRAS mutation (Supplementary Fig. S8) showing how upper and lower gastrointestinal cancers show distinct ways of altering the same oncogene (35).

An additional 14 regions of amplification were specific to EA. Six of these peaks contained genes known to contribute to cancer (CDK6, MCL1, PRKCI, MYB, MET, and FGFR2), although the others contain no previously described oncogenes. To evaluate how the large sample size enabled identification of relevant targets, we compared our analysis to the SCNA analyses of the largest previously published EA datasets, comprising 42 and 56 tumors (8, 36). Amplifications at CCNE1, MET, FGFR2, and MYB were not identified in previous datasets, but these genes have been noted to be overexpressed in EA (37–40). Many peaks that lack known oncogenes were also not noted in earlier reports. Although MET and CCNE1 amplifications had been detected previously by focused gene inquiry, MYB and FGFR2 amplifications were not noted in prior data. Our sample numbers also afforded greater resolution to identify targets of previously identified SCNAs. For example, an amplicon at 6p21.1 was reported in studies of 42 and 56 EAs to contain between 50 and 70 genes (8, 36). We narrowed this region to only 2 genes, including the vascular endothelial growth factor, VEGFA, the target of the therapeutic antibody bevacizumab. A prior study of EA also identified a 94-gene region of amplification on 3q and attributed this event to PIK3CA (36). Our analysis narrows this peak to a region containing PRKCI, >5 Mb away from PIK3CA.

The presence of FGFR2 amplifications in EA suggests a potential new therapeutic target for these tumors, similar to in GC (9). We confirmed the presence of FGFR2 amplification in individual EA cases through quantitative RT-PCR (Supplementary Fig. S9). These results indicate FGFR2 amplification may serve as a biomarker for the use of FGFR2-directed therapy in EA in addition to GC.

Four amplification peaks were restricted to gastric adenocarcinoma. Among these, only 3q27.1, containing the RTK gene, EPHB3, has been suggested to play a role in cancer progression (41). Compared with prior studies, we not only confirmed recurrent focal amplifications involving GATA4 and GATA6, but also identified novel peaks on 6p21.1 (VEGFA), 3q27.1 (EPHB3), 1p36.22, 12q15, and 1q42.3 (9).

The most significant focally amplified tyrosine kinases, ERBB2, EGFR, MET, FGFR1, and FGFR2, are known oncogenes and targets of therapeutic agents in current use or development. We detected amplifications involving one or more RTKs in 42% of EA, 28% of GC, and 14% of CRC samples (Fig. 4). Only 10% of tumors had concurrent amplifications of RTKs and KRAS.

Figure 4.

Amplified therapeutic determinants in gut adenocarcinomas. The presence of focal amplifications (inferred copy number > 2.3) is shown in red for recurrently amplified receptor tyrosine kinase genes across all tumor specimens (vertical bars).

Figure 4.

Amplified therapeutic determinants in gut adenocarcinomas. The presence of focal amplifications (inferred copy number > 2.3) is shown in red for recurrently amplified receptor tyrosine kinase genes across all tumor specimens (vertical bars).

Close modal

Finally, we compared amplifications in GI adenocarcinomas to those found in a study of 2,311 diverse cancers (12). Among the 33 focal amplifications in GI cancers, 42% overlapped with peak regions in other cancers (Supplementary Fig. S10), including 11 regions containing the known oncogenes MCL1, MYB, EGFR, FGFR1, MYC, CCND1, KRAS, MDM2, ERBB2, and CCNE1 (marked with asterisks in Fig. 3). Peak regions present in gut, but absent from nongut adenocarcinomas, encompassed genes encoding the tissue-specific transcription factors GATA4 and GATA6 and the known or putative oncogenes CDK6, FGFR2, MET, EPHB3, and EPHB6. Some of these genes are occasionally amplified in other cancer types, but did not show statistical significance. HMGA2 amplifications on chromosome 12 are notably absent in GI tumors, despite the presence of chromosome 12 amplification at MDM2 in both GI and non-GI carcinomas. Similarly, amplification of the G1–S cell-cycle–dependent kinase CDK6 is restricted to GI tumors, whereas CDK4 amplification is significant in all other tumors further suggesting that many cancer use similar pathways with multiple inputs to reach the same output (Supplementary Table S3).

Comparison of focal deletions.

We identified 16, 8, and 21 peaks of significant deletion in CRC, GC, and EA, respectively (Fig. 3B and Supplementary Fig. S11 and Table S4). Six peaks were shared across the 3 cancer types, all containing genes spanning large genomic loci (including FHIT, WWOX, and MACROD2; refs. 12, 42).

Seven deletion peaks were unique to CRC, including 2 peaks that encompass known tumor suppressors (APC and PTEN), and 5 that do not. Among the latter, a previously unidentified peak at 10q25 contains the apoptosis effector caspase gene CASP7, suggesting a mechanism to evade apoptosis (Supplementary Table S4A).

Eleven deletion peaks were unique to EA whereas none were unique to GC (Fig. 3B). Among the EA peaks, 2 contain known esophageal tumor suppressors (CDKN2A and ATM; Supplementary Table S4B and Fig. S7D). One peak contains RUNX1, which we consider further below. Two peaks were seen in both esophageal and gastric tumors, including 1 large gene (PTPRD) and 1 region (6p25.3) with no known tumor suppressor.

Among the 30 deletion peaks in the composite tumor set (Table 2), 15 were also significant in the non-GI adenocarcinoma study (Supplementary Fig. S10). The common sites included 9 peaks encompassing large genes, 4 peaks containing known deleted tumor suppressors (APC, CDKN2A, ATM, and PTEN), and 2 peaks without either. One of the latter peaks contains the effector caspase gene CASP3, suggesting that deletion of an effector caspase, CASP3 or CASP7, may mark GI adenocarcinomas more generally and not CRC alone. Focal loss of RB1 and TP53 were identified in non-GI but not in digestive tract adenocarcinomas, although 17p deletion, containing TP53, was significant in CRC and EAC. The 15 deletion peaks restricted to GI adenocarcinomas include 13 that lack known tumor suppressors (Supplementary Table S5). The other 2 contain the known tumor suppressors SMAD4 and RUNX1.

Functional significance of RUNX1 deletions in esophageal adenocarcinoma

We observed highly focal RUNX1 deletions at 21q22.12 in 15% of EAs, also noted in a recent report (Fig. 5A; ref. 36). RUNX1 behaves as a tumor suppressor in leukemia, where translocations and mutations disrupt gene function (43). We, therefore, evaluated a possible tumor suppressor function for RUNX1 in EA by reintroducing it into the EA cell line OE33, which carries a focal RUNX1 deletion (Fig. 5A). We observed a 69% reduction in anchorage-independent growth relative to GFP-infected cells (Fig. 5B and C). As we did not possess an EA cell line without deletion at the RUNX1 locus, we ectopically expressed RUNX1 in A549 lung cancer cells, which have no focal RUNX1 deletion, to evaluate for generalized cellular toxicity because of overexpression of this gene. In contrast to OE33 cells, RUNX1 expression did not significantly affect A549 colony formation (Fig. 5B and C). These results are consistent with a potential role for RUNX1 as a tumor suppressor in EA.

Figure 5.

RUNX1 reexpression can inhibit anchorage-independent growth in an esophageal adenocarcinoma cell model. A, focal deletion of locus 21q22.12 containing RUNX1 in esophageal adenocarcinomas, OE33 (EA), and A549 (non–small cell lung cancer) cell lines. B, OE33 and A549 cell lines were stably transduced with retrovirus containing RUNX1. Immunoblotting confirmed positive expression of RUNX1. C, soft agar colony formation for OE33 and A549 cells with ectopic GFP or RUNX1 expression. Representative images of colony formation at ×6.3 magnification. Quantitation of 3 independent experiments; error bars represent SEM. **, P < 0.005; n.s., not significant.

Figure 5.

RUNX1 reexpression can inhibit anchorage-independent growth in an esophageal adenocarcinoma cell model. A, focal deletion of locus 21q22.12 containing RUNX1 in esophageal adenocarcinomas, OE33 (EA), and A549 (non–small cell lung cancer) cell lines. B, OE33 and A549 cell lines were stably transduced with retrovirus containing RUNX1. Immunoblotting confirmed positive expression of RUNX1. C, soft agar colony formation for OE33 and A549 cells with ectopic GFP or RUNX1 expression. Representative images of colony formation at ×6.3 magnification. Quantitation of 3 independent experiments; error bars represent SEM. **, P < 0.005; n.s., not significant.

Close modal

These data provide the most comprehensive, high-resolution analysis to date of SCNA patterns across the 3 most common forms of gut adenocarcinoma. Our cohort size enhanced the ability to detect significant SCNAs, and we identified several focused areas of recurrent genomic alteration pointing toward genes that may contribute to cancer. Notably, the genomes of EA and GC cancers contain alterations selected for their contributions to the process of both intestinal metaplasia and malignant transformation to adenocarcinoma.

We observed more focal amplifications in upper GI adenocarcinomas compared with CRC. Unlike CRC, EA, and GC emerge in a setting of bile and acid injury, which may generate DNA strand breaks and contribute to the high rates of SCNA (44). Alternatively, distinct DNA repair pathways or selection for differing stimuli may account for these differences. It is unclear why the enhanced rates of focal amplifications in EA/GC were not matched by a similarly increased rate of focal deletions nor why the rates of deletion in GC fell below those in EA or CRC. Although, the loci of the most significant amplifications peaks fell at known or plausible oncogenes, many focal deletions peaks lie in potential fragile sites. Thus, the mechanisms and selection pressures underlying deletions may differ from those responsible for amplification.

The GATA6 and GATA4 transcription factor genes lie in the second and sixth most significant amplification peaks across the full dataset, respectively. The developmental role of these transcription factors and selective amplification in GI cancers suggests that they may add to the growing number of lineage-survival transcription factor oncogenes (45). A parallel phenomenon is amplification of SOX2 in squamous esophageal and lung carcinomas (46), an event absent in EA.

A key clinical observation emerging from these data is that focally amplified RTKs were observed most prominently in EA and GC, suggesting that genomic amplifications will be more important biomarkers in upper gastrointestinal cancers than in CRC. Recent clinical trials reveal benefit when the HER2-directed antibody trastuzumab is combined with chemotherapy in treating ERBB2-amplified or overexpressing GC/EA (11). The presence of ERBB2 amplifications in 6% of CRCs suggests that ERRB2 (HER2)-directed therapy may benefit select patients with CRC (47). In addition, based on evidence that KRAS mutation negatively predicts cetuximab response, highly prevalent KRAS amplification in upper GI tumors may similarly impact clinical decisions. When KRAS is evaluated as a biomarker in upper GI cancers, it will be important to examine both SCNAs and point mutations. Our data also point to other genomic amplifications of clinical relevance. CDK6 (48) and VEGFA focal amplifications may serve as a marker of response to targeted inhibitors. More broadly, these data point to the inclusion of gene amplifications to guide therapy in upper gastrointestinal adenocarcinomas.

Although these data suggest that many patients with EA or GC may benefit from treatment targeting an amplified RTK, it is unlikely that therapies directed against these targets alone will lead to durable responses. The progression-free survival provided by single-agent therapy with trastuzumab in GC/EA has been modest (11). The presence of complex SCNA profiles in these tumors suggest there could be co-occurring alterations that confer primary resistance (49) or enhanced genomic instability that speeds acquired resistance. In our cohort, the rates of co-occurrence between RTK-associated and other events did not significantly deviate from what would be expected for statistically independent events. However, co-occurrences were detected, and the rates with which individual pairs of events co-occur are likely to inform combinational treatment strategies. Although evaluation of RTK-targeted agents is needed, the genomic complexity and variability of these cancers suggests that combination inhibitor strategies will ultimately be essential.

Beyond these immediately clinically relevant targets, these data provide enhanced insight into specific genes responsible for different subtypes of gastrointestinal adenocarcinomas. Despite a shared intestinal origin, upper gastrointestinal cancers exhibit many distinct events from those seen in CRC, such as alteration of cell-cycle regulators (CCND1, CCNE1, CDK6, and CDKN2A). Moreover, as evidenced by the selective deletion of RUNX1 in EA, clear genomic differences exist between EA and GC. Although our analysis has helped identify loci of recurrent alteration and potential targets of these events, many recurrent regions of SCNA do not contain known oncogenes or tumor suppressors. One challenge moving forward will be to determine which of these regions harbor additional genes or noncoding elements that contribute toward intestinal metaplasia or transformation to cancer. High-resolution genome analyses of large sample numbers, as presented here, can guide future studies and inform the development of strategies to reverse the effects of the somatic genetic events that drive these cancers.

R. Beroukhim, M. Meyerson, and R.A. Shivdasani are consultants for Novartis. M. Meyerson is a founder of Foundation Medicine and Microgenomica, has commercial research grant from Novartis, is a consultant/advisory board member for Novartis and Foundation Medicine, and has patent royalties from Laboratory Corporation of America. J.D. Luketich has commercial research grant from Accuray. R.A. Shivdasani has commercial research grant from Novartis Oncology. R. Beroukhim has commercial research grant from Novartis Institutes for BioMedical Research and ownership interest (including patents) from Astrazeneca stock. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A.M. Dulak, J. van Lieshout, J. Tabernero, M. Meyerson, D.G. Beer, R. Beroukhim, A.J. Bass

Development of methodology: J. van Lieshout, Y. Imamura, J. Luketich, R. Beroukhim, A.J. Bass

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J. van Lieshout, C. Fox, S. Baca, J. Tabernero, J. Barretina, P. Enzinger, G. Corso, F. Roviello, L. Lin, S. Bandla, J. Luketich, A. Pennathur, R.A. Shivdasani, D.G. Beer, T.E. Godfrey

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A.M. Dulak, S. Schumacher, J. van Lieshout, Y. Imamura, C. Fox, B. Shim, A. Ramos, G. Saksena, J. Baselga, J. Tabernero, A. Pennathur, M. Meyerson, S. Ogino, R.A. Shivdasani, T.E. Godfrey, R. Beroukhim, A.J. Bass

Writing, review, and/or revision of the manuscript: A.M. Dulak, S. Schumacher, J. van Lieshout, B. Shim, J. Baselga, J. Tabernero, J. Barretina, P. Enzinger, G. Corso, S. Bandla, A. Pennathur, M. Meyerson, S. Ogino, R.A. Shivdasani, D.G. Beer, T.E. Godfrey, R. Beroukhim, A.J. Bass

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A.M. Dulak, J. van Lieshout, B. Shim, A. Ramos, F. Roviello, J. Luketich, S. Ogino, A.J. Bass

Study supervision: A.M. Dulak, J. Tabernero, R. Beroukhim, A.J. Bass

The authors thank the members of the Broad Institute Biological Samples Platform and Genomic Analysis Platform for their work enabling this project and also thank the physicians and hospital staff whose efforts in collecting these samples is essential to this research.

This work was supported in part by the National Institute of Health- National Cancer Institute grant 5R01CA090665 (PI: J.D. Luketich). A.J. Bass is supported by the National Cancer Institute (K08CA134931) and GI SPORE Developmental Project award—P50CA127003) and the DeGregorio Family Foundation. R. Beroukhim is supported by the National Cancer Institute (K08CA122833 and U54CA143798) and a V Foundation Scholarship. F. Roviello is supported by a grant from Istituto Toscano Tumori.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Jemal
A
,
Bray
F
,
Center
MM
,
Ferlay
J
,
Ward
E
,
Forman
D
. 
Global cancer statistics
.
CA Cancer J Clin
2011
;
61
:
69
90
.
2.
American Cancer Society
.
Available from
: http://www.cancer.org/Research/CancerFactsFigures/CancerFactsFigures/cancer-facts-figures-2011;
Vol. 1
, 
2011
.
Accessed Nov. 28, 2011
.
3.
Fearon
ER
,
Vogelstein
B
. 
A genetic model for colorectal tumorigenesis
.
Cell
1990
;
61
:
759
67
.
4.
Wood
LD
,
Parsons
DW
,
Jones
S
,
Lin
J
,
Sjoblom
T
,
Leary
RJ
, et al
The genomic landscapes of human breast and colorectal cancers
.
Science
2007
;
318
:
1108
13
.
5.
Di Nicolantonio
F
,
Martini
M
,
Molinari
F
,
Sartore-Bianchi
A
,
Arena
S
,
Saletti
P
, et al
Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer
.
J Clin Oncol
2008
;
26
:
5705
12
.
6.
Karapetis
CS
,
Khambata-Ford
S
,
Jonker
DJ
,
O'Callaghan
CJ
,
Tu
D
,
Tebbutt
NC
, et al
K-ras mutations and benefit from cetuximab in advanced colorectal cancer
.
N Engl J Med
2008
;
359
:
1757
65
.
7.
Rossi
E
,
Klersy
C
,
Manca
R
,
Zuffardi
O
,
Solcia
E
. 
Correlation between genomic alterations assessed by array comparative genomic hybridization, prognostically informative histologic subtype, stage, and patient survival in gastric cancer
.
Hum Pathol
2011
;
42
:
1937
45
.
8.
Goh
XY
,
Rees
JR
,
Paterson
AL
,
Chin
SF
,
Marioni
JC
,
Save
V
, et al
Integrative analysis of array-comparative genomic hybridisation and matched gene expression profiling data reveals novel genes with prognostic significance in oesophageal adenocarcinoma
.
Gut
2011
;
60
:
1317
26
.
9.
Deng
N
,
Goh
LK
,
Wang
H
,
Das
K
,
Tao
J
,
Tan
IB
, et al
A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets
.
Gut
2012
;
61
:
673
84
.
10.
Wainberg
ZA
,
Lin
LS
,
DiCarlo
B
,
Dao
KM
,
Patel
R
,
Park
DJ
, et al
Phase II trial of modified FOLFOX6 and erlotinib in patients with metastatic or advanced adenocarcinoma of the oesophagus and gastro-oesophageal junction
.
Br J Cancer
2011
;
105
:
760
5
.
11.
Bang
YJ
,
Van Cutsem
E
,
Feyereislova
A
,
Chung
HC
,
Shen
L
,
Sawaki
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
97
.
12.
Beroukhim
R
,
Mermel
CH
,
Porter
D
,
Wei
G
,
Raychaudhuri
S
,
Donovan
J
, et al
The landscape of somatic copy-number alteration across human cancers
.
Nature
2010
;
463
:
899
905
.
13.
The Cancer Genome Atlas Research Network
. 
Comprehensive genomic characterization defines human glioblastoma genes and core pathways
.
Nature
2008
;
455
:
1061
8
.
14.
The Cancer Genome Atlas Research Network
. 
Integrated genomic analyses of ovarian carcinoma
.
Nature
2011
;
474
:
609
15
.
15.
Mermel
CH
,
Schumacher
SE
,
Hill
B
,
Meyerson
ML
,
Beroukhim
R
,
Getz
G
. 
GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers
.
Genome Biol
2011
;
12
:
R41
.
16.
Beroukhim
R
,
Getz
G
,
Nghiemphu
L
,
Barretina
J
,
Hsueh
T
,
Linhart
D
, et al
Assessing the significance of chromosomal aberrations in cancer: methodology and application to glioma
.
Proc Natl Acad Sci U S A
2007
;
104
:
20007
12
.
17.
Bardi
G
,
Sukhikh
T
,
Pandis
N
,
Fenger
C
,
Kronborg
O
,
Heim
S
. 
Karyotypic characterization of colorectal adenocarcinomas
.
Genes Chromosomes Cancer
1995
;
12
:
97
109
.
18.
Buffart
TE
,
Louw
M
,
van Grieken
NC
,
Tijssen
M
,
Carvalho
B
,
Ylstra
B
, et al
Gastric cancers of Western European and African patients show different patterns of genomic instability
.
BMC Med Genomics
2011
;
4
:
7
.
19.
He
QJ
,
Zeng
WF
,
Sham
JS
,
Xie
D
,
Yang
XW
,
Lin
HL
, et al
Recurrent genetic alterations in 26 colorectal carcinomas and 21 adenomas from Chinese patients
.
Cancer Genet Cytogenet
2003
;
144
:
112
8
.
20.
Molkentin
JD
. 
The zinc finger-containing transcription factors GATA-4, -5, and -6. Ubiquitously expressed regulators of tissue-specific gene expression
.
J Biol Chem
2000
;
275
:
38949
52
.
21.
Muleris
M
,
Dutrillaux
AM
,
Olschwang
S
,
Salmon
RJ
,
Dutrillaux
B
. 
Predominance of normal karyotype in colorectal tumors from hereditary non-polyposis colorectal cancer patients
.
Genes Chromosomes Cancer
1995
;
14
:
223
6
.
22.
Nancarrow
DJ
,
Handoko
HY
,
Smithers
BM
,
Gotley
DC
,
Drew
PA
,
Watson
DI
, et al
Genome-wide copy number analysis in esophageal adenocarcinoma using high-density single-nucleotide polymorphism arrays
.
Cancer Res
2008
;
68
:
4163
72
.
23.
Tsafrir
D
,
Bacolod
M
,
Selvanayagam
Z
,
Tsafrir
I
,
Shia
J
,
Zeng
Z
, et al
Relationship of gene expression and chromosomal abnormalities in colorectal cancer
.
Cancer Res
2006
;
66
:
2129
37
.
24.
Baudis
M
. 
Progenetix oncogenomic.
Available from
: http://www.progenetix.net, 
2009
.
Accessed Jan. 17, 2012
.
25.
Baudis
M
,
Cleary
ML
. 
Progenetix.net: an online repository for molecular cytogenetic aberration data
.
Bioinformatics
2001
;
17
:
1228
9
.
26.
Alvarez
H
,
Opalinska
J
,
Zhou
L
,
Sohal
D
,
Fazzari
MJ
,
Yu
Y
, et al
Widespread hypomethylation occurs early and synergizes with gene amplification during esophageal carcinogenesis
.
PLoS Genet
2011
;
7
:
e1001356
.
27.
Lin
L
,
Bass
AJ
,
Lockwood
WW
,
Wang
Z
,
Silvers
AL
,
Thomas
DG
, et al
Activation of GATA binding protein 6 (GATA6) sustains oncogenic lineage-survival in esophageal adenocarcinoma
.
Proc Natl Acad Sci U S A
2012
;
109
:
4251
6
.
28.
Lin
L
,
Aggarwal
S
,
Glover
TW
,
Orringer
MB
,
Hanash
S
,
Beer
DG
. 
A minimal critical region of the 8p22-23 amplicon in esophageal adenocarcinomas defined using sequence tagged site-amplification mapping and quantitative polymerase chain reaction includes the GATA-4 gene
.
Cancer Res
2000
;
60
:
1341
7
.
29.
Smith
DI
,
Zhu
Y
,
McAvoy
S
,
Kuhn
R
. 
Common fragile sites, extremely large genes, neural development and cancer
.
Cancer Lett
2006
;
232
:
48
57
.
30.
Rothenberg
SM
,
Mohapatra
G
,
Rivera
MN
,
Winokur
D
,
Greninger
P
,
Nitta
M
, et al
A genome-wide screen for microdeletions reveals disruption of polarity complex genes in diverse human cancers
.
Cancer Res
2010
;
70
:
2158
64
.
31.
Jones
S
,
Wang
TL
,
Shih Ie
M
,
Mao
TL
,
Nakayama
K
,
Roden
R
, et al
Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma
.
Science
2010
;
330
:
228
31
.
32.
Wang
K
,
Kan
J
,
Yuen
ST
,
Shi
ST
,
Chu
KM
,
Law
S
, et al
Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer
.
Nat Genet
2011
;
43
:
1219
23
.
33.
Jang
JH
. 
Reciprocal relationship in gene expression between FGFR1 and FGFR3: implication for tumorigenesis
.
Oncogene
2005
;
24
:
945
8
.
34.
Firestein
R
,
Bass
AJ
,
Kim
SY
,
Dunn
IF
,
Silver
SJ
,
Guney
I
, et al
CDK8 is a colorectal cancer oncogene that regulates beta-catenin activity
.
Nature
2008
;
455
:
547
51
.
35.
MacConaill
LE
,
Campbell
CD
,
Kehoe
SM
,
Bass
AJ
,
Hatton
C
,
Niu
L
, et al
Profiling critical cancer gene mutations in clinical tumor samples
.
PLoS One
2009
;
4
:
e7887
.
36.
Gu
J
,
Ajani
JA
,
Hawk
ET
,
Ye
Y
,
Lee
JH
,
Bhutani
MS
, et al
Genome-wide catalogue of chromosomal aberrations in Barrett's esophagus and esophageal adenocarcinoma: a high-density single nucleotide polymorphism array analysis
.
Cancer Prev Res (Phila)
2010
;
3
:
1176
86
.
37.
Brabender
J
,
Lord
RV
,
Danenberg
KD
,
Metzger
R
,
Schneider
PM
,
Park
JM
, et al
Increased c-myb mRNA expression in Barrett's esophagus and Barrett's-associated adenocarcinoma
.
J Surg Res
2001
;
99
:
301
6
.
38.
Herrera
LJ
,
El-Hefnawy
T
,
Queiroz de Oliveira
PE
,
Raja
S
,
Finkelstein
S
,
Gooding
W
, et al
The HGF receptor c-Met is overexpressed in esophageal adenocarcinoma
.
Neoplasia
2005
;
7
:
75
84
.
39.
Iida
S
,
Katoh
O
,
Tokunaga
A
,
Terada
M
. 
Expression of fibroblast growth factor gene family and its receptor gene family in the human upper gastrointestinal tract
.
Biochem Biophys Res Commun
1994
;
199
:
1113
9
.
40.
Lin
L
,
Prescott
MS
,
Zhu
Z
,
Singh
P
,
Chun
SY
,
Kuick
RD
, et al
Identification and characterization of a 19q12 amplicon in esophageal adenocarcinomas reveals cyclin E as the best candidate gene for this amplicon
.
Cancer Res
2000
;
60
:
7021
7
.
41.
Ji
XD
,
Li
G
,
Feng
YX
,
Zhao
JS
,
Li
JJ
,
Sun
ZJ
, et al
EphB3 is overexpressed in non-small-cell lung cancer and promotes tumor metastasis by enhancing cell survival and migration
.
Cancer Res
2011
;
71
:
1156
66
.
42.
Bignell
GR
,
Greenman
CD
,
Davies
H
,
Butler
AP
,
Edkins
S
,
Andrews
JM
, et al
Signatures of mutation and selection in the cancer genome
.
Nature
2010
;
463
:
893
8
.
43.
Cohen
M
 Jr
. 
Perspectives on RUNX genes: an update
.
Am J Med Genet Part A
2009
;
149
:
2629
46
.
44.
Bernstein
H
,
Bernstein
C
,
Payne
CM
,
Dvorakova
K
,
Garewal
H
. 
Bile acids as carcinogens in human gastrointestinal cancers
.
Mutat Res
2005
;
589
:
47
65
.
45.
Garraway
LA
,
Sellers
WR
. 
Lineage dependency and lineage-survival oncogenes in human cancer
.
Nat Rev Cancer
2006
;
6
:
593
602
.
46.
Bass
AJ
,
Watanabe
H
,
Mermel
CH
,
Yu
S
,
Perner
S
,
Verhaak
RG
, et al
SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas
.
Nat Genet
2009
;
41
:
1238
42
.
47.
Bertotti
A
,
Migliardi
G
,
Galimi
F
,
Sassi
F
,
Torti
D
,
Isella
C
, et al
A molecularly annotated platform of patient-derived xenografts (“Xenopatients”) identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer
.
Cancer Disc
2011
;
1
:
508
23
.
48.
Ismail
A
,
Bandla
S
,
Reveiller
M
,
Toia
L
,
Zhou
Z
,
Gooding
WE
, et al
Early G cyclin-dependent kinases as prognostic markers and potential therapeutic targets in esophageal adenocarcinoma
.
Clin Cancer Res
2011
;
17
:
4513
22
.
49.
Maher
EA
,
Brennan
C
,
Wen
PY
,
Durso
L
,
Ligon
KL
,
Richardson
A
, et al
Marked genomic differences characterize primary and secondary glioblastoma subtypes and identify two distinct molecular and clinical secondary glioblastoma entities
.
Cancer Res
2006
;
66
:
11502
13
.

Supplementary data