Purpose:

Esophageal, gastroesophageal junction, and gastric adenocarcinoma (herein gastroesophageal adenocarcinomas) are associated with poor prognosis and limited systemic treatment options. To further understand the genomic landscape of gastroesophageal cancers and its clinical correlations, circulating tumor DNA (ctDNA) from patients’ plasma was evaluated using next-generation sequencing (NGS).

Experimental Design:

We analyzed genomic alterations of 55 patients (mostly advanced disease; 9, surgically resectable) with gastroesophageal adenocarcinomas using clinical-grade NGS performed on plasma-derived ctDNA (54–73 gene panel). The test detects single-nucleotide variants, as well as copy number amplifications, fusions, and indels in selected genes.

Results:

Seventy-six percent of patients (42/55) had ≥1 genomic alteration [including variants of unknown significance (VUS)] and 69.1% (38/55) had ≥1 characterized alteration (excluding VUSs). The median number of alterations per patient was 2 (range, 0–15). TP53 (50.9%, 28/55), PIK3CA (16.4%, 9/55), ERBB2 (14.5%, 8/55), and KRAS (14.5%, 8/55) genes were most frequently affected characterized alterations. Thirty-one patients also had tissue NGS. Concordance between tissue and ctDNA ranged from 61.3% (TP53 alterations) to 87.1% (KRAS alterations). ERBB2 alterations were significantly associated with poor overall survival (HR, 14.06; 95% confidence interval, 2.44–81.03; P = 0.003 multivariate analysis). Among patients with ≥1 alteration, no 2 patients had identical molecular portfolios. All patients with ≥1 characterized alteration had theoretically targetable alterations by an FDA-approved agent (on- or off-label). Illustrative case treated with cognate agent is presented.

Conclusions:

Evaluation of ctDNA by NGS among patients with gastroesophageal adenocarcinoma is feasible. Patients harbored heterogeneous patterns of genomics, with most having alterations that are potentially pharmacologically tractable.

Translational Relevance

Gastroesophageal adenocarcinomas are associated with poor prognosis and have limited systemic treatment options. Thus, there is an unmet need for novel diagnostic tools. Investigation of circulating tumor DNA (ctDNA) from patient plasma using clinical-grade next-generation sequencing (NGS) revealed that TP53 (50.9%, 28/55), PIK3CA (16.4%, 9/55), ERBB2 (14.5%, 8/55), and KRAS (14.5%, 8/55) were the most commonly altered genes. Tissue- and blood-derived ctDNA results were often discordant, consistent with previous work indicating intrapatient heterogeneity in gastroesophageal cancers. Presence of ERBB2 alterations was associated with significantly worse overall survival from time of ctDNA collection (HR, 14.06; 95% confidence interval, 2.44–81.03; P = 0.003 by multivariate analysis). No two patients had identical molecular portfolios, suggesting that optimal targeting with customized combination strategies may be required to control gastroesophageal cancers.

Globally, gastric and esophageal malignancies (herein gastroesophageal cancers) are one of the most frequent types of cancers with approximately 1.5 million patients diagnosed each year. They are associated with high mortality and 1.1 million patients are estimated to die each year (1). At the time of diagnosis, about 50%–60% of patients have regional lymph node involvement or distant metastatic disease (2). Combinations of systemic chemotherapies are generally used for the management of metastatic gastroesophageal cancers. Systemic therapies with antimetabolite- (5-fluorouracil or capecitabine) and platinum- (oxaliplatin or cisplatin) based therapies, as well as taxanes are widely used; however, median survival remains poor (9–11 months; refs. 3, 4). Thus, there is an urgent need to better understand the molecular biology of these neoplasms.

Along with the rapid advances in next-generation sequencing (NGS) technology, the molecular nosology of gastroesophageal cancers is now better understood. The Cancer Genome Atlas Research Network categorized patients with gastric cancer into four different subtypes: (i) Epstein–Barr virus-related group, which are associated with PIK3CA mutations, PD-L1/2 overexpression, and CDKN2A silencing; (ii) microsatellite unstable group, associated with hypermutation and MLH1 silencing; (iii) chromosomal unstable group associated with receptor tyrosine kinase and RAS activation; and (iv) genomically stable subtypes (5).

Clinically, several genomic and/or protein markers are now being used to guide treatment decisions for patients with gastroesophageal cancers. Examples include targeting of HER2 overexpressed/amplified gastric and gastroesophageal junction cancers with trastuzumab (anti-HER2 antibody; ref. 6) and PD-L1–overexpressed cases with pembrolizumab (anti-PD-1 antibody; refs. 7, 8), which are now FDA approved. Although some achievements were seen with the aforementioned targeted therapy approaches, clinical benefit has been modest (trastuzumab: 2.7 months of survival gain when added to chemotherapy; ref. 6; pembrolizumab response rate of ∼20%; ref. 7). Moreover, targeting other markers including FGFR amplification or MET overexpression, has not been able to demonstrate clinical benefit, at least as monotherapies, in the setting of gastroesophageal cancers (9, 10).

One of the major challenges to addressing genomic alterations in the clinic is tumor heterogeneity. Pectasides and colleagues compared genomic alterations between primary and metastatic lesions in 26 patients with gastroesophageal adenocarcinomas and revealed extensive differences (11). Kim and colleagues reported that more than half of the ERBB2-amplified gastroesophageal adenocarcinomas had additional oncogenic alterations, with each patient harboring unique molecular patterns that could explain the reason for the modest effects achieved with ERBB2-targeting agents (12). Moreover, dynamic change in underlying genomic alterations can evolve along with tumor progression and therapeutic pressure, which further confounds targeted therapy approaches (13).

NGS of plasma-derived circulating tumor DNA (ctDNA; also known as cell-free tumor DNA) has recently been evaluated in several tumor types (14–17). Detection of tumor-specific mutations that are shed into the blood from cancer cells can be performed on a small vial of blood, enabling the characterization of genomic alterations of tumors in a timely fashion. Although analysis of ctDNA among gastroesophageal cancers has been investigated (18), clinical application of the results is not well described. Here we investigated clinical characteristics and therapeutic outcomes among patients with gastroesophageal adenocarcinomas whose ctDNA was interrogated by clinical-grade NGS and compared the results to tissue NGS results in those patients in whom both tests were performed.

Patients

We investigated the genomic alteration status and clinical outcomes of 55 patients with gastroesophageal adenocarcinomas seen at the UC San Diego Moores Cancer Center (January 2014 to July 2017; San Diego, CA). Blood samples were evaluated at the clinical laboratory improvement amendments-licensed and College of American Pathologist-accredited clinical laboratories–Guardant Health, Inc. (http://www.guardanthealth.com) or Foundation Medicine (https://www.foundationmedicine.com) for ctDNA analysis. Tissue samples were sent to Foundation Medicine for tissue DNA testing using NGS. All investigations followed the guidelines of the UCSD Internal Review Board for data collection (NCT02478931) and for any experimental therapeutic trials for which consents were obtained.

NGS

Most ctDNA analyses were done through Guardant Health, Inc (N = 49/55). ctDNA was extracted from whole blood collected in 10 mL Streck tubes, and 5–30 ng of ctDNA was prepared for sequencing as described previously (19). All ctDNA was sequenced, including the somatic ctDNA and the germline ctDNA that is derived from natural leukocyte lysis. Germline alterations were filtered out and not reported. The fractional concentration or variant allele fraction for a given somatic mutation is calculated as the fraction of ctDNA harboring that mutation in a background of wild-type ctDNA fragments at the same nucleotide position. The analytic sensitivity reaches detection of 1–2 single mutant fragments from a 10 mL blood sample (0.1% limit of detection) and analytic specificity is greater than 99.9999%. Throughout the timeframe of this study, the ctDNA assay performed by Guardant Health, Inc. expanded from 54 to 68 to 70 to 73 genes (N = 49; Table 1; Supplementary Table S1). Degree of copy number alterations were reported as follows: 1+, 2.13–2.40, which is the 10th to 50th percentile; 2+, 2.41–4.00, which is >50th to 90th percentile; and 3+, greater than 4.0 copy numbers, which is >90th percentile. The ctDNA assay performed by Foundation Medicine interrogated 67 genes (N = 6; Table 1; Supplementary Table S2) which was described previously (20). Only nonsynonymous alterations were analyzed in this study. The variant allele fraction or fractional concentration for a given somatic mutation is derived from the fraction of ctDNA harboring that mutation in a background of wild-type ctDNA fragments at the same nucleotide position (19). When patients had multiple ctDNA evaluated at different time points, the results of ctDNA at the earlier time point was used for the analysis.

Table 1.

Patient characteristics of patients with gastroesophageal cancer who had ctDNA analysis (N = 55)

Basic characteristics (N = 55)N (%)
Age, median (range; years) 
 At diagnosis 62.6 (23.5–91.5) 
 At the time of ctDNA analysis 63.9 (24.3–91.5) 
Sex 
 Male 35 (63.6%) 
 Female 20 (36.4%) 
Ethnicity 
 Caucasian 28 (50.9%) 
 Asian 8 (14.5%) 
 Hispanic 16 (29.1%) 
 Other 3 (5.5%) 
Primary tumor location 
 Esophagus (N = 11)  
  Upper 0 (0.0%) 
  Mid 3 (5.5%) 
  Lower 8 (14.5%) 
 Gastroesophageal junction (N = 17) 17 (30.9%) 
 Stomach (N = 27)  
  Cardia/body/lesser and greater curvature 14 (25.5%) 
  Incisura/antrum/pylorus 9 (16.4%) 
  Unknown 4 (7.3%) 
Histology 
 Esophageal cancer  
  Moderately differentiated 6 (10.9%) 
  Poorly differentiated 5 (9.1%) 
 Gastroesophageal junction cancer  
  Moderately differentiated 3 (5.5%) 
  Poorly differentiated 7 (12.7%) 
  Signet ring cells 3 (5.5%) 
  Unknown 4 (7.3%) 
 Gastric cancer  
  Moderately differentiated 3 (5.5%) 
  Poorly differentiated 13 (23.6%) 
  Signet ring cells 10 (18.2%) 
  Unknown 1 (1.8%) 
Disease status at the time of ctDNA analysis 
 Metastatic or recurrent 44 (80.0%) 
 Locally advanced 2 (3.6%) 
 Surgically resectablea 9 (16.4%) 
Technique of ctDNA analysis 
 Foundation Medicine (panel of 67 genes) 6 (10.9%) 
 Guardant, Inc. 49 (89.1%) 
  Panel of 54 genes 1 (1.8%) 
  Panel of 68 genes 8 (14.5%) 
  Panel of 70 genes 26 (47.3%) 
  Panel of 73 genes 14 (25.5%) 
Median number of alterations per patient (range)b 2 (0–15) 
Median number of characterized alteration per patient (range) 1 (0–7) 
Number of patients with ≥1 alteration (including VUS) 42 (76.4%) 
Number of patients with ≥1 characterized alteration 38 (69.1%) 
Number of patients with ≥5% of allele frequency 13 (23.6%) 
Median of highest allele frequency (range; %) 0.8 (0–50.7) 
Number of patients who also had tissue NGS 31 (56.4%) 
Basic characteristics (N = 55)N (%)
Age, median (range; years) 
 At diagnosis 62.6 (23.5–91.5) 
 At the time of ctDNA analysis 63.9 (24.3–91.5) 
Sex 
 Male 35 (63.6%) 
 Female 20 (36.4%) 
Ethnicity 
 Caucasian 28 (50.9%) 
 Asian 8 (14.5%) 
 Hispanic 16 (29.1%) 
 Other 3 (5.5%) 
Primary tumor location 
 Esophagus (N = 11)  
  Upper 0 (0.0%) 
  Mid 3 (5.5%) 
  Lower 8 (14.5%) 
 Gastroesophageal junction (N = 17) 17 (30.9%) 
 Stomach (N = 27)  
  Cardia/body/lesser and greater curvature 14 (25.5%) 
  Incisura/antrum/pylorus 9 (16.4%) 
  Unknown 4 (7.3%) 
Histology 
 Esophageal cancer  
  Moderately differentiated 6 (10.9%) 
  Poorly differentiated 5 (9.1%) 
 Gastroesophageal junction cancer  
  Moderately differentiated 3 (5.5%) 
  Poorly differentiated 7 (12.7%) 
  Signet ring cells 3 (5.5%) 
  Unknown 4 (7.3%) 
 Gastric cancer  
  Moderately differentiated 3 (5.5%) 
  Poorly differentiated 13 (23.6%) 
  Signet ring cells 10 (18.2%) 
  Unknown 1 (1.8%) 
Disease status at the time of ctDNA analysis 
 Metastatic or recurrent 44 (80.0%) 
 Locally advanced 2 (3.6%) 
 Surgically resectablea 9 (16.4%) 
Technique of ctDNA analysis 
 Foundation Medicine (panel of 67 genes) 6 (10.9%) 
 Guardant, Inc. 49 (89.1%) 
  Panel of 54 genes 1 (1.8%) 
  Panel of 68 genes 8 (14.5%) 
  Panel of 70 genes 26 (47.3%) 
  Panel of 73 genes 14 (25.5%) 
Median number of alterations per patient (range)b 2 (0–15) 
Median number of characterized alteration per patient (range) 1 (0–7) 
Number of patients with ≥1 alteration (including VUS) 42 (76.4%) 
Number of patients with ≥1 characterized alteration 38 (69.1%) 
Number of patients with ≥5% of allele frequency 13 (23.6%) 
Median of highest allele frequency (range; %) 0.8 (0–50.7) 
Number of patients who also had tissue NGS 31 (56.4%) 

aSurgically resectable cases (N = 9) indicates that ctDNA analysis was done prior to surgery, except for 1 case whose analysis was done 3 weeks after the surgery. All N = 9 patients had R0 resection (clear margins).

bIncludes characterized alterations and VUSs.

Thirty-one patients had both ctDNA and tissue DNA analysis. All tissue DNA analyses were performed by Foundation Medicine as described previously (21, 22). The assay for tissue DNA was designed to include all genes known to be somatically altered in human solid tumors that are validated targets for therapy and interrogated 236 genes, as well as 47 introns of 19 genes commonly rearranged in cancer (n = 2) and 315 genes, as well as introns of 28 genes commonly rearranged in cancer (N = 29).

Endpoints, statistical methods, and case studies

Descriptive statistics were used to summarize the genomic alterations identified in this study. The Mann–Whitney U test was used for continuous data and Fisher exact test was used for categorical data. Concordances of ctDNA and tissue DNA were quantified by concordance percentage and Kappa value with the SE. Kappa values are interpreted by commonly used agreement categories: κ = 1 (perfect agreement) to κ = 0 (no agreement other than would be expected by chance). Overall survival (OS) was calculated from the time of ctDNA analysis to last follow-up. Survival analyses were assessed by Kaplan–Meier analysis and Cox proportional hazard model was used to estimate HRs with 95% confidence intervals (CI). For multivariate analysis, variables with P < 0.20 in univariate analysis were included in the multivariate regression model.

Patient characteristics among patients with gastroesophageal cancer evaluated for ctDNA

A total of 55 patients with gastroesophageal adenocarcinomas were evaluated for ctDNA. Tumors located in esophagus, gastroesophageal junction, and stomach were represented as follows: 20.0% (N = 11), 30.9% (N = 17), and 49.1% (n = 27), respectively (Table 1; Fig. 1). Among all patients with gastroesophageal adenocarcinomas (N = 55), the median age at the time of diagnosis was 62.6 years old (range, 23.5–91.5), and 63.6% (N = 35) were men. ctDNA tests were performed prior to surgical resection in 9 patients (16.4%). Among patients who had ctDNA analysis, tissue NGS was performed in 31 patients (56.4%; Table 1).

Figure 1.

Consort diagram of patients with esophageal, gastroesophageal, and gastric cancer who had ctDNA analysis (N = 55). ctDNA, circulating tumor DNA; GE, gastroesophageal; NGS, next-generation sequencing; PREDICT study, Profile Related Evidence Determining Individualized Cancer Therapy study.

Figure 1.

Consort diagram of patients with esophageal, gastroesophageal, and gastric cancer who had ctDNA analysis (N = 55). ctDNA, circulating tumor DNA; GE, gastroesophageal; NGS, next-generation sequencing; PREDICT study, Profile Related Evidence Determining Individualized Cancer Therapy study.

Close modal

Among these 55 patients evaluated for ctDNA, 76.4% (N = 42) had at least one genomic alteration [includes characterized alterations and variants of unknown significance (VUS)] and 69.1% (N = 38) had at least one characterized alteration. The median number of alterations per patient was 2 (range: 0–15); the median number of characterized alterations per patient was 1 (range: 0–7). The most common characterized alteration was TP53 (50.9%, N = 28) followed by PIK3CA (16.4%, N = 9), ERBB2 (14.5%, N = 8), and KRAS (14.5%, N = 8; Fig. 2; Supplementary Table S3). Thirteen patients (23.6%) had ≥5% of allele frequency and the median percentage of highest allele frequency was 0.8% (range: 0%–50.7%; Table 1).

Figure 2.

Frequency of genomic alterations by ctDNA analysis among patients with gastroesophageal cancers (N = 55). Frequency represents percent of patients with an alteration. The most common alteration was in the TP53 gene (50.9% of patients, N = 28) followed by the ERBB2 (18.1%, N = 10), PIK3CA (16.3%, N = 9), and KRAS genes (14.5%, N = 8). See Supplementary Table S3. VUS, variants of unknown significance.

Figure 2.

Frequency of genomic alterations by ctDNA analysis among patients with gastroesophageal cancers (N = 55). Frequency represents percent of patients with an alteration. The most common alteration was in the TP53 gene (50.9% of patients, N = 28) followed by the ERBB2 (18.1%, N = 10), PIK3CA (16.3%, N = 9), and KRAS genes (14.5%, N = 8). See Supplementary Table S3. VUS, variants of unknown significance.

Close modal

Number of alterations with possible cognate-targeted therapies

Among 55 cases with gastroesophageal cancers, a total of 159 alterations were identified (including VUSs). Among those alterations, 68.6% (109/159) were characterized alterations, including substitutions [51.6% (82/159)] and amplifications [17.0% (27/159)]. Among the 109 characterized alterations, 89.9% (98/109) were potentially targetable with FDA-approved agents as on- or off-label use, and an additional 8.2% (8/109) were theoretically targetable with therapies that are currently in clinical trials. Altogether, among all characterized alterations, 97.2% (106/109) were theoretically actionable either with agents that are approved by the FDA (including off-label) or with agents that are in clinical trials. Among all 55 patients with gastroesophageal cancers, 69.1% (N = 38; all patients with ≥1 characterized alteration) had ≥1 theoretically actionable alterations by an FDA-approved agent (on- or off-label). Patients who did not have targetable alterations were those who only had VUSs (7.3%, N = 4) or patients without detectable alteration (23.6%, N = 13).

Distinctness of genomic alterations among 55 patients with gastroesophageal cancers

Among the 38 patients who had ≥1 characterized alteration, no 2 patients had identical molecular portfolios (e.g., TP53 R273H and TP53 R175H considered as molecularly distinct), while 8 patients had identical genomic portfolios (e.g., TP53 R273H and TP53 R175H considered as genomically identical; ID#15, 27, 29, 31, 40, 48, 51; Supplementary Table S4).

Concordance between ctDNA and tissue DNA testing

Of 31 patients who had both ctDNA and tissue DNA analyses, the median time interval between tissue and ctDNA collection was 1.3 months (range 0–47.1 months). The overall concordance rate was 61.3% for TP53, 83.9% for ERBB2, 74.2% for PIK3CA, and 87.1% for KRAS alterations (statistically significant concordance only seen with ERBB2 alterations (P = 0.048; Supplementary Table S5A). When overall concordance was compared between ctDNA and tissue NGS, 1 patient [3.2% (1/31)] had complete concordance (TP53 R342*, PTEN H75fs*2, and KRAS amplification were found in both ctDNA and tissue; Supplementary Table S4; ID: 43). Partial concordance (e.g., ERBB2 amplification found in both ctDNA and tissue NGS, but TP53 alteration only found in tissue NGS) was seen in 54.8% (17/31) of cases. There was no concordance seen in 41.9% (13/31) of cases (including 10 patients without detectable characterized alterations from ctDNA; Supplementary Table S4). Similar concordance was observed when the interval between blood draw and tissue biopsy was ≤6 months (N = 19 patients) versus >6 months (N = 12 patients), but the small number of individuals in each group precludes definitive conclusions (Supplementary Table S5B). When concordance was evaluated depending on the site of biopsies [primary site (N = 23) vs. metastatic site (N = 8)], higher concordance was observed between ctDNA and metastatic sites for TP53 and ERBB2 alterations (concordance rate between ctDNA and primary site vs. ctDNA and metastatic site was as follows: TP53: 52.2% vs. 87.5%, ERBB2: 78.3% vs. 100%); however, these differences were not statistically significant (Supplementary Table S5C). Because treatment can affect ctDNA levels, we ran the concordance analysis of the 20 patients who had not received systemic therapy or were off treatment for at least 4 weeks. We found similar concordance rates to those in the analysis of all patients (Supplementary Table S5D).

Survival analysis among 46 patients with locally advanced, metastatic, or recurrent gastroesophageal cancer

Patients with locally advanced, metastatic or recurrent gastroesophageal cancers (N = 46) were included in the survival analysis. Characteristics with at least 5 patients affected were included. When OS was evaluated from the time of ctDNA analysis to last follow up, in the univariate analysis, gender, PIK3CA, and KRAS alterations, as well as lines of systemic therapies (≥2 lines) before the ctDNA analysis were not statistically associated with the OS outcome. On the other hand, patients with TP53 alterations, highest %ctDNA ≥ 1.65% and cumulative total %ctDNA ≥ 2.3% had a trend towards worse OS (HR: 2.28; 95% CI: 0.72–7.29, P = 0.154; HR: 2.49, 95% CI: 0.78–8.00, P = 0.115; and HR: 2.50, 95% CI: 0.75–8.28, P = 0.122, respectively; cutoff of %ctDNA ≥ 1.65% and ≥2.3% was chosen because it was the median %ctDNA). Patients whose ctDNA was obtained ≥6.8 months from the time of metastatic/recurrent disease had a trend toward better OS (HR, 0.48; 95% CI, 0.15–1.46; P = 0.183; cutoff of 6.8 months was chosen because it was the median). ERBB2 alterations were significantly associated with worse OS (HR, 8.02; 95% CI, 2.41–26.69; P < 0.001) in univariate analysis (Table 2; Fig. 3). After the multivariate analysis, ERBB2 alterations continued to be an independent factor associated with poor OS (HR, 14.06; 95% CI, 2.44–81.03; P = 0.003). Patients whose ctDNA were obtained ≥6.8 months from the time of metastatic/recurrent disease was associated with better OS (HR, 0.18; 95% CI, 0.04–0.82; P = 0.026; Table 2).

Table 2.

OS from the time of ctDNA analysis among patients with locally advanced, metastatic, or recurrent gastroesophageal cancers (N = 46)

Univariate analysisMultivariate analysisa
CharacteristicsMedian OS (months)HR (95% CI)PHR (95% CI)P
OS from the time of ctDNA analysis (months) 
Age at the diagnosis 
 ≥63 (n = 22) vs. not (n = 24) 11.5 vs. 25.1 2.19 (0.74–6.46) 0.148 1.82 (0.53–6.30) 0.344 
Gender 
 Men (n = 31) vs. Women (n = 15) 20.2 vs. 14.8 0.64 (0.23–1.78) 0.392 — — 
Genomic alterationsb 
TP53 (n = 26) vs. not (n = 20) 20.2 vs. 25.1 2.28 (0.72–7.29) 0.154 1.19 (0.21–6.73) 0.845 
ERBB2 (n = 8) vs. not (n = 38) 1.7 vs. 20.2 8.02 (2.41–26.69) <0.001 14.06 (2.44–81.03) 0.003 
PIK3CA (n = 8) vs. not (n = 38) NR vs. 14.8 1.71 (0.47–6.21) 0.413 — — 
KRAS (n = 8) vs. not (n = 38) 14.8 vs. 20.2 1.60 (0.44–5.83) 0.472 — — 
Highest %ctDNAc 
 ≥1.65% (n = 23) vs. not (n = 23) NR vs. 20.2 2.49 (0.78–8.00) 0.115 1.04 (0.17–6.33) 0.968 
Total %ctDNAd 
 ≥2.3% (n = 24) vs. not (n = 22) 14.8 vs. 20.2 2.50 (0.75–8.28) 0.122 0.94 (0.10–9.20) 0.957 
Lines of systemic therapy prior to ctDNA analysis (received ≥2 lines) 
 Yes (n = 15) vs. No (n = 31) 20.2 vs. 14.8 0.83 (0.26–2.68) 0.751 — — 
Time from metastatic/recurrent disease to ctDNA collection ≥6.8 monthse 
 Yes (n = 23) vs. No (n = 23) 20.2 vs. 11.5 0.48 (0.15–1.46) 0.183 0.18 (0.04–0.82) 0.026 
Univariate analysisMultivariate analysisa
CharacteristicsMedian OS (months)HR (95% CI)PHR (95% CI)P
OS from the time of ctDNA analysis (months) 
Age at the diagnosis 
 ≥63 (n = 22) vs. not (n = 24) 11.5 vs. 25.1 2.19 (0.74–6.46) 0.148 1.82 (0.53–6.30) 0.344 
Gender 
 Men (n = 31) vs. Women (n = 15) 20.2 vs. 14.8 0.64 (0.23–1.78) 0.392 — — 
Genomic alterationsb 
TP53 (n = 26) vs. not (n = 20) 20.2 vs. 25.1 2.28 (0.72–7.29) 0.154 1.19 (0.21–6.73) 0.845 
ERBB2 (n = 8) vs. not (n = 38) 1.7 vs. 20.2 8.02 (2.41–26.69) <0.001 14.06 (2.44–81.03) 0.003 
PIK3CA (n = 8) vs. not (n = 38) NR vs. 14.8 1.71 (0.47–6.21) 0.413 — — 
KRAS (n = 8) vs. not (n = 38) 14.8 vs. 20.2 1.60 (0.44–5.83) 0.472 — — 
Highest %ctDNAc 
 ≥1.65% (n = 23) vs. not (n = 23) NR vs. 20.2 2.49 (0.78–8.00) 0.115 1.04 (0.17–6.33) 0.968 
Total %ctDNAd 
 ≥2.3% (n = 24) vs. not (n = 22) 14.8 vs. 20.2 2.50 (0.75–8.28) 0.122 0.94 (0.10–9.20) 0.957 
Lines of systemic therapy prior to ctDNA analysis (received ≥2 lines) 
 Yes (n = 15) vs. No (n = 31) 20.2 vs. 14.8 0.83 (0.26–2.68) 0.751 — — 
Time from metastatic/recurrent disease to ctDNA collection ≥6.8 monthse 
 Yes (n = 23) vs. No (n = 23) 20.2 vs. 11.5 0.48 (0.15–1.46) 0.183 0.18 (0.04–0.82) 0.026 

Abbreviations: NR, not reached

aVariables with P < 0.20 in univariate analysis (log-rank test) were included in multivariate analysis.

bIncluded only characterized alterations (no VUSs); only included characterized alterations seen in >5 patients

cUsed the median of highest %ctDNA value as a cutoff.

dTotal %DNA refers to the sum of allele frequency detected in each patient. Used the medial of total %ctDNA value as a cutoff.

eTime point of 6.8 months was chosen because it was the median time from metastatic/recurrent disease to ctDNA collection.

Figure 3.

Kaplan–Meier curves for OS from the time of ctDNA analysis to last follow-up depending on the ERBB2 alteration status (N = 46; 9 patients were excluded because they had resectable disease for which surgery was performed). Patients with ERBB2 alterations had significantly worse OS compared with patients without ERBB2 alteration (HR, 8.02; 95% CI, 2.41–26.69; P < 0.001; univariate analysis).

Figure 3.

Kaplan–Meier curves for OS from the time of ctDNA analysis to last follow-up depending on the ERBB2 alteration status (N = 46; 9 patients were excluded because they had resectable disease for which surgery was performed). Patients with ERBB2 alterations had significantly worse OS compared with patients without ERBB2 alteration (HR, 8.02; 95% CI, 2.41–26.69; P < 0.001; univariate analysis).

Close modal

Detection of ctDNA among patients with early-stage and advanced-stage gastroesophageal cancers

Among 9 patients whose ctDNA was examined prior to resection, 3 (33.3%) had detectable characterized alterations, while 76.1% (35/46) of patients with metastatic, recurrent, or locally advanced diseases had detectable alterations (P = 0.019). Among patients who underwent surgery (N = 9), the median number of alteration (0 vs. 3, P = 0.009) and median value of highest allele frequency of %ctDNA (0 vs. 1.7, P = 0.001) were significantly lower than that of patients with metastatic, recurrent, or locally advanced diseases (Supplementary Table S6).

Representative case among patients who received matched targeted therapies based on ctDNA results

Among 55 patients evaluated for ctDNA, 24 patients received systemic therapy after the ctDNA analysis. Among those 24 patients, 70.8% (17/24) had at least one actionable genomic alteration (the remaining 7 patients had no characterized alterations). However only 1 of 24 patients (4.2%) was treated with matched targeted therapy approach (Fig. 1; Supplementary Table S7).

The summary of the patient who received matched targeted therapy is given in Supplementary Table S7.

Patient ID.42.

This is a 68-year-old woman with gastroesophageal junction adenocarcinoma who was found to have multiple liver metastases and lymphadenopathy at diagnosis. Both ctDNA and tissue NGS revealed EGFR amplification. The patient was started on dual anti-EGFR therapy (ref. 23; cetuximab and erlotinib) and achieved partial response [67% decrease by RECIST 1.1, progression-free survival (PFS) of 18 months; ref. 17]. Along with radiographic response (Fig. 4A), serial ctDNA analysis 4 and 12 months after the first ctDNA analysis showed normalization of EGFR copy number in plasma (pretreatment EGFR copy number = 143, down to reference range; Fig. 4B).

Figure 4.

Representative case of gastroesophageal cancer that was managed with matched targeted therapy approach. A and B, 68-year-old female with gastroesophageal junction adenocarcinoma with multiple liver metastases and lymphadenopathy (A, left). ctDNA-revealed EGFR amplification. On the basis of the ctDNA analysis, patient was started on dual anti-EGFR therapy (cetuximab and erlotinib; ref. 23). Patient was also given nivolumab based on positive PD-L1 by IHC (8); however, drug was held after one dose due to grade III rash (patient continued to receive dual anti-EGFR therapy). After 3 months of therapy, patient achieved partial response [A, middle; best response = 67% decrease (A, right), PFS = 18 months] along with the normalization of EGFR copy number in blood circulation 4 and 12 months after the first ctDNA analysis (B).

Figure 4.

Representative case of gastroesophageal cancer that was managed with matched targeted therapy approach. A and B, 68-year-old female with gastroesophageal junction adenocarcinoma with multiple liver metastases and lymphadenopathy (A, left). ctDNA-revealed EGFR amplification. On the basis of the ctDNA analysis, patient was started on dual anti-EGFR therapy (cetuximab and erlotinib; ref. 23). Patient was also given nivolumab based on positive PD-L1 by IHC (8); however, drug was held after one dose due to grade III rash (patient continued to receive dual anti-EGFR therapy). After 3 months of therapy, patient achieved partial response [A, middle; best response = 67% decrease (A, right), PFS = 18 months] along with the normalization of EGFR copy number in blood circulation 4 and 12 months after the first ctDNA analysis (B).

Close modal

Patients with advanced gastroesophageal cancers have a poor prognosis with a median survival of 9–11 months (3, 4). Although molecular characteristics of gastroesophageal cancers have been investigated (5), to date, understanding clinical correlates of genomic data and capitalizing on this information in the patient care setting has been limited (6, 7, 9, 10). Herein, we report the biologic and clinical correlates of genomic alterations among 55 patients with mostly advanced-stage gastroesophageal cancers using blood-derived ctDNA interrogated by clinical-grade NGS.

Altogether, 76.4% (42/55) of patients had at least one nonsynonymous alteration detected from ctDNA and 69.1% (38/55) had at least one characterized alteration (VUSs excluded). The most frequent characterized alterations were in the TP53 gene (50.9%, 28/55) followed by the PIK3CA (16.4%, 9/55), ERBB2 (14.5%, 8/55), and KRAS genes (14.5%, 8/55; Fig. 2; Supplementary Table S3), which is consistent with a previous report (18). Among frequently altered genes, concordance rate between tissue DNA and ctDNA varied from 61%–87% depending on the alterations (Supplementary Table S5A). This observation is consistent with a previous report by Pectasides and colleagues, which showed discordance between tumor DNA and ctDNA in gastroesophageal cancers (11). This may not be surprising because genomic alterations from the areas of cancer that were not biopsied (e.g., distant metastases) or intratumoral heterogeneity are likely being uncovered by ctDNA analysis. Indeed, Pectasides and colleagues noted discordance in findings between primary tumors and metastases in gastroesophageal cancers (11). In our study, comparison between the blood draw and tissue biopsy ≤ 6 months apart (N = 19) versus >6 months apart (N = 12) did not reveal a difference in concordance rate among frequently altered genes (Supplementary Table S5B). These observations differ from previous reports in nongastric cancers that showed high concordance between tissue DNA and blood-derived ctDNA when the interval between two tests was short (24, 25). The relatively small number of patients in our study and in previous work (11) may have confounded the results and/or the concordance in gastric cancers may be lower than in other types of malignancies. Technical differences between the tissue and blood ctDNA assays cannot be ruled out.

Interestingly, patients with early-stage/surgically resectable disease had a significantly lower number of alterations detected from ctDNA, as well as lower variant allele fraction of ctDNA when compared with patients with metastatic/advanced disease (Supplementary Table S6). This observation is consistent with previous reports that showed that the level of ctDNA was associated with underlying tumor burden and can demonstrate dynamic changes along with the therapeutic course (24, 26). Moreover, among patients with early-stage colon cancer, postsurgical detection of ctDNA was strongly associated with tumor recurrence suggesting that ctDNA may be used as a surrogate for minimal residual disease after surgery (27). Further investigation in this respect is necessary in patients with gastroesophageal cancers. In regard to outcome, prior reports have shown an association between high %ctDNA and survival (24, 25). In this study, we found that high %ctDNA (evaluated via the ctDNA alteration presenting the highest variant allele fraction, as well as by calculating the cumulative percentages of all ctDNAs in each patient) showed a trend toward correlation with OS in univariate, but not in multivariate, analysis (Table 2).

Notably, among several alterations that were identified, presence of an ERBB2 alteration was significantly associated with poor OS (HR, 14.06; 95% CI, 2.44–81.03; P = 0.003 by multivariate analysis; ERBB2 altered vs. not; Table 2; Fig. 3). Trastuzumab is approved for gastroesophageal cancers with HER2 overexpression or amplification; addition of trastuzumab to chemotherapy demonstrated clinical benefit (trastuzumab plus chemotherapy vs. chemotherapy alone: median OS: 13.8 vs. 11.1 months; ToGA trial; P = 0.0046; ref. 6). The survival in our patients with ERBB2-altered malignancies was considerably shorter than in the ToGA trial, perhaps because the ToGA trial included only patients who had not had prior therapy in the metastatic setting. Also none of our patients were treated with ErbB2-targeting agents after the ctDNA analysis. In addition, detection of ERBB2 overexpression or amplification can differ depending on the various methodologies used (Supplementary Table S8), which can confound the clinical outcome. Previous reports indicate that gastroesophageal cancers with ERBB2 alterations commonly harbor genomic coalterations that can potentially drive therapeutic resistance to anti-HER2–directed therapies (12). In this study, we have also observed that all patients with ERBB2 alterations (N = 8) harbored at least one coalteration, including anomalies in FGFR2, RAF1, PIK3CA, and KRAS that can be associated with resistance to anti-HER2 regimens (Supplementary Table S4; patient ID: 1, 4, 17, 30, 32, 33, 47, and 53). These observations suggest that cotargeting of resistance signals may enhance the efficacy of Erbb2-antagonist agents. Furthermore, among patients whose tumors harbored ≥1 characterized alteration, no 2 patients had identical molecular portfolios (Supplementary Table S4). Studies with customized combination regimens have been initiated (28).

Although there is growing evidence that matched targeted therapy is potentially promising as a therapeutic approach (29–32), one of the realistic challenges to this strategy has been the low rate of patients receiving matched treatments (∼5%–20% in many studies; refs. 29, 33–35). Low target–drug matching rate is often due to a number of reasons, including lack of drug accessibility, deterioration of patient's condition at the time of matching, or lack of actionable targets (36). In this study, we have also observed that molecularly matched therapies based upon profiling with ctDNA was underutilized in the clinic, as only 1.8% (1/55) of patients were treated in this manner (Fig. 1; Supplementary Table S7) despite the fact 69.1% (38/55) of patients had theoretically tractable targets (Supplementary Table S4). Although there was only 1 patient who received matched cognate agents, this individual showed clinical benefit, with a partial response (67% regression) and PFS of 18 months in a patient with EGFR amplification who was given dual anti-EGFR therapies (cetuximab and erlotinib; Fig. 4; Supplementary Table S7). Further clinical investigation is warranted.

There were several limitations to this study. First, the study was performed retrospectively with a small sample size at a single institution. Second, not all patients had tissue NGS as a comparator. Third, it is conceivable that using different ctDNA assays may provide different results, even if the assays are clinical grade. Fourth, the number of genes evaluated changed over time (from 54 to 68 to 70 to 73 gene panels for the Guardant assay), which makes direct comparison challenging; however, it should be noted that only the genes found in both the tissue and the ctDNA panel were compared for concordance. Fifth, some of the concordance was driven by the wild-type cases (where both ctDNA and tissue would be negative for an alteration); hence, the P values might be misleading for positive concordance. Sixth, different systemic therapies at the time of ctDNA analysis can affect the level of variant allele fractions, which may have confounded the survival analysis. Finally, results of germline alterations were filtered out and not reported in this study. To comprehensively understand the biology of cancer in each patient, evaluation of both germline, as well as somatic alterations may be required. Thus, the current findings require further validation with larger numbers of patients in the setting of prospective studies using newer methodologies, as they become available, because the capability to detect alterations is improving with time.

In conclusion, we have evaluated 55 patients with gastroesophageal cancers who had blood-derived ctDNA analysis by clinical-grade NGS. The most frequent alterations were in TP53 (50.9%) followed by PIK3CA (16.4%), ERBB2 (14.5%), and KRAS genes (14.5%). At least one alteration was identified in 76.4% of patients. Concordance between ctDNA and tissue DNA among commonly altered genes ranged from 61.3% to 87.1%. Discordant results may be due to the dynamic changes in ctDNA after treatment or tumor progression, tumor heterogeneity, or from disparities in sensitivity between tissue and ctDNA analysis. Technical issues cannot be ruled out. ERBB2 alterations were associated with significantly worse OS (median OS = 1.7 vs. 20.2 months). Importantly, among patients who had at least one characterized alteration, no 2 patients had identical molecular portfolios, suggesting that customized therapy may be necessary. Although the number of patient who received therapy that matched ctDNA analysis was small, the patient showed benefit. Further investigations of the clinical utility of blood-derived ctDNA among patients with gastroesophageal cancers are needed.

J.K. Sicklick is a consultant/advisory board member for CarsGen, Grand Rounds, Loxo Oncology, and reports receiving commercial research support from Foundation Medicine and Novartis Pharmaceuticals. R. Kurzrock has ownership interests (including patents) at IDbyDNA anD CureMatch Inc., is a consultant/advisory board member for Actuate Therapeutics, LOXO, NeoMed, Roche, and X-Biotech, and reports receiving commercial research grants from Foundation Medicine, Genentech, Guardant Health, Incyte, Konica, Merck Serono, Pfizer, and Sequenom. No potential conflicts of interest were disclosed by the other authors.

Conception and design: S. Kato, R. Okamura, P.T. Fanta, R. Kurzrock

Development of methodology: S. Kato, R. Okamura, R. Kurzrock

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Kato, R. Okamura, J.M. Baumgartner, H. Patel, K. Kelly, P.T. Fanta

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Kato, R. Okamura, H. Patel, P.T. Fanta, S.M. Lippman, R. Kurzrock

Writing, review, and/or revision of the manuscript: S. Kato, R. Okamura, J.M. Baumgartner, H. Patel, L. Leichman, K. Kelly, J.K. Sicklick, S.M. Lippman, R. Kurzrock

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S. Kato, R. Okamura, S.M. Lippman

Study supervision: R. Okamura, S.M. Lippman, R. Kurzrock

Funded in part by the Joan and Irwin Jacobs Fund (to R. Kurzrock) and the Jon Schneider Memorial Cancer Research Fund (to J.K. Sicklick, P.T. Fanta), as well as by National Cancer Institute grants P30 CA023100 (to R. Kurzrock), K08 CA168999, and R21 CA192072 (all to J.K. Sicklick).

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.
Howlader
N
,
Noone
AM
,
Krapcho
M
,
Miller
D
,
Bishop
K
,
Kosary
CL
, et al
SEER Cancer Statistics Review, 1975–2014
.
Bethesda, MD
:
NCI
; 
2017
. Available from: https://seer.cancer.gov/csr/1975_2014/
3.
Cunningham
D
,
Starling
N
,
Rao
S
,
Iveson
T
,
Nicolson
M
,
Coxon
F
, et al
Capecitabine and oxaliplatin for advanced esophagogastric cancer
.
N Engl J Med
2008
;
358
:
36
46
.
4.
Van Cutsem
E
,
Moiseyenko
VM
,
Tjulandin
S
,
Majlis
A
,
Constenla
M
,
Boni
C
, et al
Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 Study Group
.
J Clin Oncol
2006
;
24
:
4991
7
.
5.
Cancer Genome Atlas Research Network
. 
Comprehensive molecular characterization of gastric adenocarcinoma
.
Nature
2014
;
513
:
202
9
.
6.
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
.
7.
Fuchs
CS
,
Doi
T
,
Jang
RW-J
,
Muro
K
,
Satoh
T
,
Machado
M
, et al
KEYNOTE-059 cohort 1: efficacy and safety of pembrolizumab (pembro) monotherapy in patients with previously treated advanced gastric cancer
.
J Clin Oncol
35
:
15s
, 
2017
(suppl; abstr 4003)
8.
Patel
SP
,
Kurzrock
R
. 
PD-L1 expression as a predictive biomarker in cancer immunotherapy
.
Mol Cancer Ther
2015
;
14
:
847
56
.
9.
Catenacci
DVT
,
Tebbutt
NC
,
Davidenko
I
,
Murad
AM
,
Al-Batran
SE
,
Ilson
DH
, et al
Rilotumumab plus epirubicin, cisplatin, and capecitabine as first-line therapy in advanced MET-positive gastric or gastro-oesophageal junction cancer (RILOMET-1): a randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet Oncol
2017
;
18
:
1467
82
.
10.
Van Cutsem
E
,
Bang
YJ
,
Mansoor
W
,
Petty
RD
,
Chao
Y
,
Cunningham
D
, et al
A randomized, open-label study of the efficacy and safety of AZD4547 monotherapy versus paclitaxel for the treatment of advanced gastric adenocarcinoma with FGFR2 polysomy or gene amplification
.
Ann Oncol
2017
;
28
:
1316
24
.
11.
Pectasides
E
,
Stachler
MD
,
Derks
S
,
Liu
Y
,
Maron
S
,
Islam
M
, et al
Genomic heterogeneity as a barrier to precision medicine in gastroesophageal adenocarcinoma
.
Cancer Discov
2018
;
8
:
37
48
.
12.
Kim
J
,
Fox
C
,
Peng
S
,
Pusung
M
,
Pectasides
E
,
Matthee
E
, et al
Preexisting oncogenic events impact trastuzumab sensitivity in ERBB2-amplified gastroesophageal adenocarcinoma
.
J Clin Invest
2014
;
124
:
5145
58
.
13.
Hiley
C
,
de Bruin
EC
,
McGranahan
N
,
Swanton
C
. 
Deciphering intratumor heterogeneity and temporal acquisition of driver events to refine precision medicine
.
Genome Biol
2014
;
15
:
453
.
14.
Kato
S
,
Krishnamurthy
N
,
Banks
KC
,
De
P
,
Williams
K
,
Williams
C
, et al
Utility of genomic analysis in circulating tumor DNA from patients with carcinoma of unknown primary
.
Cancer Res
2017
;
77
:
4238
46
.
15.
Riviere
P
,
Fanta
PT
,
Ikeda
S
,
Baumgartner
J
,
Heestand
GM
,
Kurzrock
R
. 
The mutational landscape of gastrointestinal malignancies as reflected by circulating tumor DNA
.
Mol Cancer Ther
2018
;
17
:
297
305
.
16.
Schrock
AB
,
Pavlick
D
,
Klempner
SJ
,
Chung
JH
,
Forcier
B
,
Welsh
A
, et al
Hybrid capture-based genomic profiling of circulating tumor DNA from patients with advanced cancers of the gastrointestinal tract or anus
.
Clin Cancer Res
2018
;
24
:
1881
90
.
17.
Schwaederle
M
,
Chattopadhyay
R
,
Kato
S
,
Fanta
PT
,
Banks
KC
,
Choi
IS
, et al
Genomic alterations in circulating tumor DNA from diverse cancer patients identified by next-generation sequencing
.
Cancer Res
2017
;
77
:
5419
27
.
18.
Catenacci
DVT
,
Nagy
RJ
,
Braiteh
FS
,
Kim
S
,
Kwak
EL
,
Maron
SB
, et al
Cell free circulating tumor DNA (ctDNA) landscape in patients with advanced gastroesophageal adenocarcinoma (GEC)
.
J Clin Oncol
35
:
15s
, 
2017
(suppl; abstr 47).
19.
Lanman
RB
,
Mortimer
SA
,
Zill
OA
,
Sebisanovic
D
,
Lopez
R
,
Blau
S
, et al
Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumor DNA
.
PLoS One
2015
;
10
:
e0140712
.
20.
Ou
SI
,
Young
L
,
Schrock
AB
,
Johnson
A
,
Klempner
SJ
,
Zhu
VW
, et al
Emergence of preexisting MET Y1230C mutation as a resistance mechanism to crizotinib in NSCLC with MET exon 14 skipping
.
J Thorac Oncol
2017
;
12
:
137
40
.
21.
Frampton
GM
,
Fichtenholtz
A
,
Otto
GA
,
Wang
K
,
Downing
SR
,
He
J
, et al
Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing
.
Nat Biotechnol
2013
;
31
:
1023
31
.
22.
Thomas
RK
,
Nickerson
E
,
Simons
JF
,
Janne
PA
,
Tengs
T
,
Yuza
Y
, et al
Sensitive mutation detection in heterogeneous cancer specimens by massively parallel picoliter reactor sequencing
.
Nat Med
2006
;
12
:
852
5
.
23.
Maron
SB
,
Alpert
L
,
Kwak
HA
,
Lomnicki
S
,
Chase
L
,
Xu
D
, et al
Targeted therapies for targeted populations: anti-EGFR treatment for EGFR amplified gastroesophageal adenocarcinoma
.
Cancer Discov
2018
;
8
:
696
713
.
24.
Schwaederle
M
,
Husain
H
,
Fanta
PT
,
Piccioni
DE
,
Kesari
S
,
Schwab
RB
, et al
Use of liquid biopsies in clinical oncology: pilot experience in 168 patients
.
Clin Cancer Res
2016
;
22
:
5497
505
.
25.
Schwaederle
MC
,
Patel
SP
,
Husain
H
,
Ikeda
M
,
Lanman
RB
,
Banks
KC
, et al
Utility of genomic assessment of blood-derived circulating tumor DNA (ctDNA) in patients with advanced lung adenocarcinoma
.
Clin Cancer Res
2017
;
23
:
5101
11
.
26.
Thierry
A
,
El Messaoudi
S
,
Mollevi
C
,
Raoul
J
,
Guimbaud
R
,
Pezet
D
, et al
Clinical utility of circulating DNA analysis for rapid detection of actionable mutations to select metastatic colorectal patients for anti-EGFR treatment
.
Ann Oncol
2017
;
28
:
2149
59
.
27.
Tie
J
,
Wang
Y
,
Tomasetti
C
,
Li
L
,
Springer
S
,
Kinde
I
, et al
Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer
.
Sci Transl Med
2016
;
8
:
346ra92
.
28.
Sicklick
JK
,
Leyland-Jones
B
,
Kato
S
,
Hahn
M
,
Williams
C
,
De
P
, et al
Personalized, molecularly matched combination therapies for treatment-na
.
J Clin Oncol
35
:
15s
, 
2017
(suppl; abstr 2512).
29.
Tsimberidou
AM
,
Iskander
NG
,
Hong
DS
,
Wheler
JJ
,
Falchook
GS
,
Fu
S
, et al
Personalized medicine in a phase I clinical trials program: the MD Anderson Cancer Center initiative
.
Clin Cancer Res
2012
;
18
:
6373
83
.
30.
Schwaederle
M
,
Zhao
M
,
Lee
JJ
,
Lazar
V
,
Leyland-Jones
B
,
Schilsky
RL
, et al
Association of biomarker-based treatment strategies with response rates and progression-free survival in refractory malignant neoplasms: a meta-analysis
.
JAMA Oncol
2016
;
2
:
1452
9
.
31.
Jardim
DL
,
Schwaederle
M
,
Wei
C
,
Lee
JJ
,
Hong
DS
,
Eggermont
AM
, et al
Impact of a biomarker-based strategy on oncology drug development: a meta-analysis of clinical trials leading to FDA approval
.
J Natl Cancer Inst
2015
;
107
:
djv253
.
32.
Schwaederle
M
,
Zhao
M
,
Lee
JJ
,
Eggermont
AM
,
Schilsky
RL
,
Mendelsohn
J
, et al
Impact of precision medicine in diverse cancers: a meta-analysis of phase II clinical trials
.
J Clin Oncol
2015
;
33
:
3817
25
.
33.
Stockley
TL
,
Oza
AM
,
Berman
HK
,
Leighl
NB
,
Knox
JJ
,
Shepherd
FA
, et al
Molecular profiling of advanced solid tumors and patient outcomes with genotype-matched clinical trials: the Princess Margaret IMPACT/COMPACT trial
.
Genome Med
2016
;
8
:
109
.
34.
Lopez-Chavez
A
,
Thomas
A
,
Rajan
A
,
Raffeld
M
,
Morrow
B
,
Kelly
R
, et al
Molecular profiling and targeted therapy for advanced thoracic malignancies: a biomarker-derived, multiarm, multihistology phase II basket trial
.
J Clin Oncol
2015
;
33
:
1000
7
.
35.
Andre
F
,
Bachelot
T
,
Commo
F
,
Campone
M
,
Arnedos
M
,
Dieras
V
, et al
Comparative genomic hybridisation array and DNA sequencing to direct treatment of metastatic breast cancer: a multicentre, prospective trial (SAFIR01/UNICANCER)
.
Lancet Oncol
2014
;
15
:
267
74
.
36.
Lewin
J
,
Siu
LL
. 
Cancer genomics: the challenge of drug accessibility
.
Curr Opin Oncol
2015
;
27
:
250
7
.

Supplementary data