Open Access

Genetic profiling of somatic alterations by Oncomine Focus Assay in Korean patients with advanced gastric cancer

  • Authors:
    • Joonhong Park
    • Sang-Il Lee
    • Soyoung Shin
    • Jang Hee Hong
    • Han Mo Yoo
    • Jeong Goo Kim
  • View Affiliations

  • Published online on: August 20, 2020     https://doi.org/10.3892/ol.2020.11990
  • Article Number: 129
  • Copyright: © Park et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer is one of the leading causes of cancer‑associated death; however, analysis of its molecular and clinical characteristics has been complicated by its histological and etiological heterogeneity. The present study aimed to estimate somatic mutation profiling in gastric cancer. To do so, targeted next‑generation sequencing (NGS) was performed with the Oncomine Focus Assay to compare the clinicopathological characteristics with the mutation profiles in 50 patients with advanced gastric cancer (AGC). Among the 35 hotspot genes and 19 genes for copy number variations (CNVs), 18 single nucleotide variants (SNVs) or small insertions and deletions (14 missense and four frameshift mutations), and 10 amplifications were identified. To examine the association between mutation profiles and clinicopathological characteristics, each element of the clinicopathological characteristics was categorized into three groups: No alteration, PI3K catalytic subunit α (PIK3CA) alterations and alterations other than PIK3CA. Fisher's exact test identified no statistical differences between the clinicopathological characteristics, with the exception of the Tumor‑Node‑Metastasis (TNM) T stage between the three groups. Cases of AGC with somatic alterations but no PIK3CA exhibited a significant difference in the TNM T stage compared with those with no alterations or PIK3CA alterations (P=0.044). In addition, AGC with PIK3CA alterations was categorized by Lauren's classification to the intestinal type only. The distribution of Lauren's classification in AGC with PIK3CA alterations was statistically different compared with AGC with alterations other than PIK3CA (P=0.028), but not compared with AGC with no alterations (P=0.076). In conclusion, the present study demonstrated a molecular profiling approach that identified potential molecular classifications for gastric cancer and suggested a framework for precision medicine in AGC.

Introduction

Gastric cancer is a major cause of cancer-associated mortality. It ranks as the 5th most common neoplasm and the 3rd most deadly cancer, and was responsible for >1,000,000 new cases with an estimated 783,000 deaths in 2018 (1). However, analysis of the clinical and genetic characteristics of gastric cancer has been complicated by its etiological and histological heterogeneity (2). Etiologically, gastric cancer is often accompanied by infectious agents such as Helicobacter pylori or Epstein-Barr virus, susceptible genetic variants and environmental factors, along with the accumulation of epigenetic and genetic changes (3). Histopathological classification systems such as the Lauren's classification and World Health Organization classification system (4) have limited clinical usefulness as to which classifications unify a clinical correlation with a high validity and practicability in diagnosis and prognostic outcome, making the development of relevant classifiers that can help patient care an urgent priority (5). Thus, molecular classification of gastric cancer has been developed, and candidate drivers and dysregulated pathways of notable subtypes of gastric cancer have been identified (6). Several molecular targeted therapies associated with survival outcomes in other cancer types are currently in clinical research for the treatment of gastric cancer, including the inhibitors of epidermal growth factor receptor (EGFR), fibroblast growth factor receptor (FGFR), Met proto-oncogene receptor tyrosine kinase (MET), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) and vascular endothelial growth factor (VEGF) (7).

The application of next-generation sequencing (NGS) technologies exploiting whole genome sequencing to targeted sequencing has served an important role in the identification of genetic variations and anomalies in patients with gastric cancer, which has improved our understanding of the molecular profiles and heterogeneity of gastric cancers (1,2). Targeted NGS represents a resource- and cost-efficient approach, enabling the detection of somatic alterations of potential interest. The Oncomine Focus Assay (OFA) is a targeted NGS assay for the simultaneous and rapid identification of single nucleotide variants (SNVs), short insertions and deletions (indels; 35 genes), copy number variations (CNVs; 19 genes) and gene rearrangements (23 genes) in 52 cancer genes with therapeutic relevance, and can detect potential targets and current actionable genetic variants for personalized medicine (8). The OFA is designed for use with the Ion Torrent Personal Genome Machine (PGM™) that generates 10–100 Mb pairs (Mbp) of sequence data on various chips within several hours of instrument run time and leverages the uniquely minimal total of DNA or RNA input (10 ng), which is useful for frequent analysis of small amounts of clinical samples (9). Combined with Ion AmpliSeq technology, this approach enables highly accurate and reproducible sequence analysis of various types of tumor specimens (10).

The present study aimed to compare the clinicopathological characteristics with the mutation profiles of 50 Korean patients with advanced gastric cancer (AGC) by targeted NGS assay along with the OFA panel.

Materials and methods

DNA isolation and quantification

The study protocol was approved by the Institutional Review Board of The Catholic University of Korea (approval no. DC18SESI0113). All subjects provided written informed consent for clinical and molecular analyses and publication before the study. A total of 50 patients with AGC who received surgical resection between January 2015 and February 2019 at the Department of Surgery, Chungnam National University Hospital (Daejeon, Republic of Korea) were enrolled in the present study. The cohort comprised of 72% (36/50) male and 28% (14/50) female Korean patients with a mean age of 66 years (age range, 39–91 years). Genomic DNA was isolated from 50 frozen human AGC tissues using the RecoverAll Total Nucleic Acid Isolation kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Amplifiable genomic DNA was determined by fluorometric quantitation using Qubit 2.0 Fluorometer with Qubit dsDNA HS Assay kits and the TaqMan RNase P Detection Reagents kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols and was considered appropriate when the nucleic acid concentration was >30 ng/µl.

Library preparation

DNA libraries were constructed using the Ion AmpliSeq™ Library kit 2.0 (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The Oncomine Focus DNA Assay (Thermo Fisher Scientific, Inc.) was used to generate sequencing libraries from 10 ng input genomic DNA per specimen. The OFA panel can identify hotspot mutations, including SNVs, indels (35 genes) and CNVs (19 genes) that are commonly implicated in human cancers and relevant to targeted treatment of solid tumors (9). Unique Ion Xpress Barcode 1–16 and Ion P1 Adapter (Thermo Fisher Scientific, Inc.) were ligated to the amplicons and subsequently purified to ensure that each individual sample had a unique ID. The final amplicon libraries were amplified, purified and equalized to ~100 pM using an AMPure XP Reagent (Beckman Coulter, Inc.).

Semiconductor sequencing

A total of six uniquely barcoded library samples were pooled for sequencing per run on an Ion 318™ v2 chip (Thermo Fisher Scientific, Inc.). The Ion Chef™ System (Thermo Fisher Scientific, Inc.) was applied using the Ion PGM™ Hi-Q™ Chef Kit for fully automated template preparation and Ion 318™ v2 chip loading. Single-end sequence analysis was performed using the Ion PGM™ Hi-Q™ Sequencing Kit on the Ion Torrent PGM™ (Thermo Fisher Scientific, Inc.) for 200 base-read sequencing.

Variant calling and data analysis

Raw data from the DNA panel was generated for sequence reads, collected, processed and trimmed using the Ion Torrent platform-specific pipeline software as follows. Removal of polyclonal and poor signal profile reads as well as 3′ quality trimming of reads was performed using Torrent Suite Assay Development Mode v5.0 (Thermo Fisher Scientific, Inc.). Reads were aligned to the human genome hg19 (https://www.ncbi.nlm.nih.gov/assembly/GCF_000001405.13/) and Ion Reporter v5.1 software package (Thermo Fisher Scientific, Inc.) was used for data analysis of the DNA panel. A cut off of 500× coverage was applied to all analyses in the present study; the target regions with >500× demonstrated sufficient and uniform amplification and sequencing coverage, with mutant alleles detected at >5% allele frequency. Briefly, the ‘Oncomine Focus-520-w2.4-DNA-Single Sample’ automatic workflow in Ion Reporter was used to identify and annotate the SNVs, indels and CNVs from the OFA with the following Torrent Variant Caller parameter settings: Frequency cutoff for supporting a variant, SNV 0.04, indel 0.07, Hotspot 0.03; total coverage required of reads or no-call, SNV 15, indel 15, Hotspot 15; proportion of variant alleles coming overwhelmingly from one strand, SNV 0.96, indel 0.9, Hotspot 0.96 for SNV and indel calls; and median of the absolute values of all pairwise differences <0.4; 5% confidence interval CNV ploidy ≥ gain of 2 over normal for CNV calling.

Candidate variant prioritization

Pathogenic impact of missense mutations other than hotspot mutations on gene function was estimated using in silico prediction tools such as ‘Damaging’ (score 0) by SIFT (11) and ‘Probably damaging’ (score >0.8) by Polyphen-2 (12). Conservation change of an affected amino acid was compared by aligning protein sequences of various vertebrate species obtained from the Evolutionary Annotation Database (http://www.h-invitational.jp/evola/). In addition, the candidate mutation was investigated whether it has been reported as pathogenic for gastric cancer in the sequence databases including COSMIC (https://cancer.sanger.ac.uk/cosmic) (13) or ClinVar (https://www.ncbi.nlm.nih.gov/clinvar/).

Immunohistochemistry (IHC) for CDK4, EGFR, ERBB2, FGFR2, KRAS, MET, MYC and PIK3CA

A total of 10 tissue samples with the gene amplification identified by the OFA assay were fixed in buffered 10% formalin at 65°C for 10 min and embedded in paraffin. Formalin-fixed, paraffin-embedded (FFPE) samples were sectioned at a thickness of 4 µm. The BenchMark XT automated slide processing system (Ventana Medical Systems, Inc.) was used for deparaffinization pretreatment with Cell Conditioning 1 solution (Ventana) and ultraviolet irradiation to abrogate endogenous hydroperoxidase activity according to the manufacturer's instructions. These sections were incubated at 37°C for 24 min with primary antibodies (1:100; Abcam) against CDK4 (cat. no. ab108357), EGFR (cat. no. ab52894), ERBB2 (cat. no. ab16662), FGFR2 (cat. no. ab58201), KRAS (cat. no. ab180772), MET (cat. no. ab216574), MYC (cat. no. ab32072), PIK3CA (cat. no. ab40776). Sections, were then incubated with horseradish peroxidase-conjugated goat anti-rabbit IgG heavy & light chain secondary antibody (Abcam) at 37°C for 10 min. Sections were counterstained with hematoxylin II (Ventana) for 5 min and bluing reagent (Ventana) for 5 min at 37°C. Slides were imaged under a light microscope (BX51; Olympus Corporation). The intensity of immunostaining for protein expression was scored as follows: 0, negative; 1+, weak; 2+, moderate; and 3+, strong in >10% of tumor cells; only 2+ or 3+ were interpreted as being positive as previously described (14).

Statistical analysis

Data were presented as the means ± standard deviation. Statistical analysis was performed using MedCalc Statistical Software Version 17.6 (MedCalc Software, Ltd.). Normality was assessed using Kolmogorov-Smirnov and Shapiro-Wilk tests, and one-way analysis of variance followed by Tukey's post hoc test was used to compare the means of age between three groups categorized by Lauren's classification or mutation profiles by targeted NGS. The Fisher's exact test was used to compare the clinicopathological characteristics and mutation profiles between two or three groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Mutation analysis

A median sequencing coverage depth of 1,845× (range, 129–2,000×) was achieved for the 50 gastric cancer tissues. Integrative analysis using the Ion Reporter identified somatic mutations with allele frequencies between 6 and 47% in tumor DNA samples without matched normal controls, in which a variant was classified as germline origin if its mutation frequency was near 50% (heterozygous) or 100% (homozygous), or otherwise classified as somatic. Null mutations (nonsense, frameshift or canonical ±1 or 2 splice sites) and missense variants with allele frequency <0.00001 predicted to be deleterious or damaging that were not registered in COSMIC database were also included. After applying stringent parameters for reliable variant calling (coverage depth >500×; allele frequency >5%) by filtering out unlikely pathogenic variants or potential raw base calling errors, at least one somatic alteration including SNVs, indels and amplification was detected in 22/50 (44%) patients. Details of the somatic alteration profiles identified by targeted NGS in these 22 cases of AGC are summarized in Tables I and II. Of the 35 hotspot genes and 19 genes for copy number variations (CNVs), 18 single nucleotide variants (SNVs) or small indels (14 missense and four frameshift mutations) and 10 amplifications were identified (Fig. 1). Amplification of the CDK4, EGFR, ERBB2, FGFR2, KRAS, MET, MYC and PIK3CA genes was confirmed to be graded as ≥2 by additional IHC (Fig. 2). Somatic alterations in the PIK3CA gene were the most frequently identified genetic alteration, occurring in 8/22 (36%) samples, followed by four ERBB2, four KRAS, two EGFR, two FGFR3 and one each for the other tested genes (Fig. 3).

Table I.

Results of somatic alteration profiles identified by the Oncomine focus assay in 22 patients with advanced gastric cancer.

Table I.

Results of somatic alteration profiles identified by the Oncomine focus assay in 22 patients with advanced gastric cancer.

SampleGeneTranscriptBase changeCodon changeEffectFreq %COSMIC IDdbSNP
CN03ERBB2NM_004448.3c.2524G>Ap.Val842IleMissense10COSM14065rs1057519738
CN11EGFRNM_005228.4c.2227G>Ap.Ala743ThrMissense22rs759256622
CN11KRASNM_033360.3c.38G>Ap.Gly13AspMissense47COSM532rs112445441
CN13FGFR3NM_000142.4c.274delCp.Gln92Serfs*6Frameshift28
CN14ALKNM_004304.4c.4061G>Tp.Cys1354PheMissense16rs963770969
CN15PIK3CANM_006218.3 c.328_330delGAAp.Glu109delFrameshift41COSM24710rs1060500031
CN16PIK3CANM_006218.3c.323G>Ap.Arg108HisMissense27COSM27497rs886042002
CN23PIK3CANM_006218.3 c.328_330delGAAp.Glu109delFrameshift20COSM24710
CN27ERBB2NM_004448.3c.2033G>Ap.Arg678GlnMissense9COSM436498rs1057519862
CN28PIK3CANM_006218.3c.1633G>Ap.Glu545LysMissense8COSM763rs104886003
CN34CTNNB1NM_001904.3c.98C>Tp.Ser33PheMissense6COSM5669rs121913400
CN38FGFR1NM_001174067.1c.2359C>Tp.Arg787CysMissense20
CN40KRASNM_033360.3c.34G>Ap.Gly12SerMissense24COSM517rs121913530
PIK3CANM_006218.3c.1633G>Ap.Glu545LysMissense7COSM763rs104886003
CN44FGFR3NM_000142.4c.274delCp.Gln92Serfs*6Frameshift18
PIK3CANM_006218.3c.1390T>Gp.Ser464AlaMissense39
CN46PIK3CANM_006218.3c.1633G>Ap.Glu545LysMissense23COSM763rs104886003
CN49BRAFNM_004333.4c.1780G>Ap.Asp594AsnMissense6COSM27639rs397516896

[i] dbSNP, database for single nucleotide polymorphisms; freq, mutation frequency.

Table II.

Results of copy number variations identified by the Oncomine Focus DNA Assay in 22 patients with advanced gastric cancer.

Table II.

Results of copy number variations identified by the Oncomine Focus DNA Assay in 22 patients with advanced gastric cancer.

SampleGeneLength, kbVariant classCytoBand
CN02MYC4.4Amplification 8q24.21(128,748,885-128,753,261)×13.51
CN14ERBB215.1Amplification 17q12(37,868,126-37,883,249)×24.58
CN16ERBB215.1Amplification 17q12(37,868,126-37,883,249)×14.25
CN17FGFR2107.0Amplification 10q26.13(123,247,505-123,354,466)×13.78
CN18PIK3CA35.4Amplification 3q26.32(178,916,683-178,952,097)×13.23
CN20KRAS35.5Amplification 12p12.1(25,364,761-25,400,274)×19.43
CN30CDK44.0Amplification 12q14.1(58,142,052-58,146,026)×14.39
CN38MET121.0Amplification 7q31.2(116,313,480-116,434,565)×9.82
CN41EGFR60.6Amplification 7p11.2(55,198,956-55,259,538)×15.35
CN42KRAS35.5Amplification 12p12.1(25,364,761-25,400,274)×14.69
Comparison of clinicopathological characteristics according to Lauren's classification

The 50 AGC cases were categorized into three subtypes according to Lauren's classification: Diffuse, intestinal and mixed type, and the clinicopathological characteristics and mutation profiles of patients in these three groups were compared. The mixed type was more frequently associated with a younger age compared with the diffuse or intestinal type (P=0.003; Table III). By contrast, the intestinal type more frequently exhibited moderate differentiation compared with the diffuse or mixed type (P<0.001). The frequency of mutations identified by targeted NGS with the OFA was not statistically different among the three groups (P=0.240).

Table III.

Comparison of clinicopathologic findings according to Lauren's classification subtypes in 50 patients with advanced gastric cancer.

Table III.

Comparison of clinicopathologic findings according to Lauren's classification subtypes in 50 patients with advanced gastric cancer.

CharacteristicDiffuse (n=15)Intestinal (n=30)Mixed (n=5)P-value
Male8 (53%)24 (80%)4 (80%)0.138
Age, years (range)62 (39–91)71 (52–89)56 (53–59)0.003
Differentiation <0.001
  Moderate0 (0%)24 (80%)1 (20%)
  Poor15 (100%)6 (20%)4 (80%)
TNM T stage 0.830
  21 (7%)3 (10%)0 (0%)
  33 (20%)9 (30%)1 (20%)
  4a11 (73%)18 (60%)4 (80%)
TNM N stage 0.378
  03 (20%)7 (23%)0 (0%)
  11 (7%)9 (30%)1 (20%)
  24 (27%)7 (23%)1 (20%)
  3a5 (33%)7 (23%)2 (40%)
  3b2 (13%)0 (0%)1 (20%)
TNM M stage 0.088
  014 (93%)30 (100%)4 (80%)
  11 (7%)0 (0%)1 (20%)
Any mutations (SNV + indel) 0.240
  No13 (87%)19 (63%)3 (60%)
  Yes2 (13%)11 (37%)2 (40%)
Any amplifications 1.000
  No12 (80%)24 (80%)4 (80%)
  Yes3 (20%)6 (20%)1 (20%)
Any mutations or amplifications 0.522
  No10 (67%)16 (53%)2 (40%)
  Yes5 (33%)14 (47%)3 (60%)

[i] SNV, single nucleotide variant; indel, small insertion, duplication and deletion; TNM, Tumor-Node-Metastasis (4).

Comparison of clinicopathological characteristics according to mutation profiles

To examine the association between mutation profiles and clinicopathological characteristics, each element in the clinicopathological characteristics was categorized into three groups: No alteration, PIK3CA alterations and alterations other than PIK3CA. By Fisher's exact test, there were no statistical differences between clinicopathological findings except TNM staging (T) between the three groups (Table IV). AGCs with somatic alterations but no PIK3CA showed statistical difference in TNM staging (T), compared to AGCs without or with PIK3CA alterations (P=0.044). In addition, AGC with the PIK3CA alterations was categorized by Lauren's classification to the intestinal type only. The distribution of Lauren's classification in AGC with PIK3CA alterations was statistically different compared with AGC with alterations other than PIK3CA (P=0.028), but not with AGC with no alterations (P=0.076).

Table IV.

Comparison of clinicopathologic characteristics according to mutation profiles identified by Oncomine focus assay.

Table IV.

Comparison of clinicopathologic characteristics according to mutation profiles identified by Oncomine focus assay.

CharacteristicsNo alterations (n=28)Other alterationsa (n=14)PIK3CA alterations (n=8)P-value
Male19 (68%)9 (64%)8 (100%)0.152
Age, years (range)62 (39–84)64 (53–91)77 (52–89)0.156
Differentiation 0.675
  Moderate14 (50%)6 (43%)5 (63%)
  Poor14 (50%)8 (57%)3 (37%)
TNM T stage 0.044
  21 (4%)3 (21%)0 (0%)
  311 (39%)1 (7%)1 (12%)
  4a16 (57%)10 (72%)7 (88%)
TNM N stage 0.892
  05 (18%)3 (21%)2 (25%)
  16 (21%)3 (21%)2 (25%)
  25 (18%)5 (37%)2 (25%)
  3a/10 (36%)2 (14%)2 (25%)
  3b2 (7%)1 (7%)0 (0%)
TNM M stage 0.702
  027 (96%)13 (93%)8 (100%)
  11 (4%)1 (7%)0 (0%)
Lauren's classification 0.028b
0.076c
  Diffuse10 (36%)5 (36%)0 (0%)
  Intestinal16 (57%)6 (43%)8 (100%)
  Mixed2 (7%)3 (21%)0 (0%)

a Alternations other than PIK3CA (single nucleotide variant, small insertion, duplication, deletion and amplification) identified by the Oncomine focus assay in the present study.

b The distribution of Lauren's classification in AGC with PIK3CA alterations vs. AGC with alterations other than PIK3CA.

c GC with PIK3CA alterations vs. AGC with no alterations. PIK3CA, PI3K catalytic subunit α; Lauren's classification (4); TNM, Tumor-Node-Metastasis (4).

Discussion

Molecular characterization of gastric cancer may offer new tools for effective therapeutic strategies for well-defined sets of patients, as well as new clinical trial designs leading to an improvement of medical management of this disease (15). These novel classifications allow the identification of relevant gastric cancer genomic subsets by using techniques such as genomic screening, functional studies and molecular or epigenetic characterization (16). The large scale study of molecular profiling on gastric cancer in The Cancer Genome Atlas (TCGA), including a report from TCGA (5) and an independent study from the Asian Cancer Research Group (17), provide an outstanding opportunity to establish advanced molecular classifiers and predictors for the diagnosis and treatment of gastric cancer. In addition to these studies, several smaller studies have performed NGS to thoroughly establish the genomics of gastric cancer (1821). Similar to these small-scale studies, the results of the present study demonstrated that AGC with PIK3CA alterations was associated with the intestinal type in Lauren's classification. The PIK3CA mutations activate the PI3K/Akt signaling pathway, have been reported in several types of carcinoma and are associated with negative outcome (22). PIK3CA amplification is associated with increased Akt phosphorylation levels, suggesting that this genetic alteration may serve a significant role in activating the PI3K/Akt signaling pathway that contributes to gastric carcinogenesis (23). Kim et al (24) have suggested that PIK3CA-mutated gastric cancer is a distinct disease entity that may require a different therapeutic approach. PIK3CA mutations were associated with Akt activation and high tumor aggressiveness in gastric cancer (24). In addition, high PIK3CA expression was significantly associated with tumor invasiveness, phenotype and poor patient survival (25). Unlike previous studies using quantitative PCR (24) or IHC (25) for the PIK3CA alterations only, the present study confirmed that PIK3CA mutation and amplification in gastric cancer were associated with adverse clinical manifestation using multi-gene analysis. The results of the present gene panel study demonstrated that AGC with mutated PIK3CA tended to be of an advanced TNM T stage (T4a, 88%), compared with AGC with wild-type PIK3CA (57%) or with mutations other than PIK3CA (72%), although Epstein-Barr virus (EBV) in situ hybridization was not investigated; a previous study demonstrated that PIK3CA mutations were more dispersed in EBV-positive cancer, but localized in the kinase domain (exon 20) in EBV-negative cancer (5).

The plethora of data obtained from recent NGS studies has resulted in the discovery of other candidate genes with similar functions to those of CDH1 and TP53 as classic driver genes of gastric cancer that may have valuable influence on therapeutic decisions and clinical outcomes (6). The novel main categories of driver mutations that have been ascertained by NGS include cell motility/cytoskeleton (26), chromatin remodeling (27), receptor tyrosine kinase pathway genes (28) and Wnt signaling (29). A recent study using NGS demonstrated notable mutation distributions in seven candidate genes (A-kinase anchoring protein 6, cyclic nucleotide binding domain containing 1, collagen type XIV alpha 1 chain, -box and WD repeat domain containing 7, integrin subunit alpha V, neurobeachin and xin actin binding repeat containing 2) that had not been previously report to be prominently mutated in gastric cancer (30). For medical genetic testing, which is crucial for precision medicine in cancer treatment, target NGS with a gene panel is advantageous due to cost savings, enhanced depth of coverage and precise target enhancement (31). Therefore, clinically helpful molecular classification based on targeted sequencing with a gene panel may enable the use of precise medicine in gastric cancer (21,30,32).

The identification of specific cancer subgroups is also enabling precise selection of patients who are likely to respond to immunotherapy (33). Through conventional methods for EBV and microsatellite instability, as well as the use of emerging genetics testing that focuses on a gene panel for mutations and amplifications, the proposed genetic group may be applied to new cases of AGC (5). Tumor heterogeneity and the incomplete understanding of the complex tumor biology represent an obstacle to the overcoming of the ‘one size fits all’ era of gastric cancer treatment (33). The most disturbed pathways in gastric cancer include adherens junction and focal adhesion (18). The clustered mutations in recurrent hotspots influence the functional domain and produce defective RHOA signaling, facilitating escape from anoikis in organoid cultures (18). In addition, gastric cancers with different Lauren's classifications exhibit diverse characteristics, and EGF containing fibulin extracellular matrix protein 1 (EFEMP1), frizzled related protein (FRZB) and keratin 23 (KRT23) have been identified as prognostic factors for gastric cancer subtypes (34). EFEMP1 and FRZB may be involved in diffuse gastric cancer-specific pathways, such as cell adhesion; KRT23 may serve a critical role in intestinal gastric cancer, considering that it has been demonstrated to be an oncogene that can influence DNA damage and proliferation response of colon cancer cells (35).

There were several limitations to the present study. The most notable limitation was the small sample size, as it was difficult to investigate significant relationships for the genetic landscape of gastric cancers from the present data. Thus, it is essential to further improve the molecular characterization of gastric cancer subtypes in order to provide researchers and medical oncologists with new tools for patient selection and stratification in future clinical development programs and subsequent trials (36). Comprehensive large-scale studies on the molecular classification in gastric cancer covering recent genomic, transcriptomic, proteomic and epigenomic features are required (37). Another limitation of the present study was that due to the inherent problems with the OFA, particularly important variants may have not been called; the OFA does not identify the mutations of previously known such as AT-rich interaction domain 1A (ARID1A), cadherin 1 (CDH1) and tumor protein p53 (TP53) as well as new, such as catenin alpha 2, GLI family zinc finger 3, mucin 6, and ring finger protein 43) significantly mutated driver genes. The OFA applied in the present study was a relatively small gene panel to be used for identification of complicated genetic alterations in AGC. Although PIK3CA gene alterations were the most frequently identified in the present study, frequencies of genetic alteration in ARID1A, LDL receptor related protein 1B and TP53 are higher compared with PIK3CA in public cancer genome databases such as cBioPortal for Cancer Genomics (www.cbioportal.org). Since PIK3CA alterations are significantly enriched in EBV-positive gastric cancer samples (28), EBV in situ hybridization should be required in a future study to confirm whether the previously published data may be extrapolated to the cohort of the present study. Similarly, although 15 diffuse type gastric cancer samples were included in the present study, no CDH1 mutations were reported, as the OFA did not cover coding region of the CDH1 gene. The identification of CDH1 gene mutations in diffuse type gastric cancer is important since most diffuse type gastric cancers are known to harbor pathogenic CDH1 mutations.

In conclusion, the present study demonstrated a molecular profiling approach that identified the potential molecular classifications for gastric cancer and suggested a framework for precision medicine in AGC. The improvements in this field may influence the discovery of novel driver mutations as well as sophisticated classification systems for gastric cancer that may be crucial for its pathogenesis if they can be effectively applied to improve the clinical outcome and therapeutic paradigm of AGC.

Acknowledgements

The authors would like to thank Mr. Taekyu Lee (Thermo Fisher Scientific, Inc.) for their contribution in providing technical support for Ion Torrent next generation sequencing.

Funding

This work was supported by The Catholic University of Korea Daejeon St. Mary's Hospital, Clinical Research Institute Grant (grant nos. CMCDJ-P-2018-010 and CMCDJ-P-2019-012) and the Chungnam National University Hospital Research Fund 2009. The bio-specimens and data used for this study were provided by the Biobank of Chungnam National University Hospital (Daejeon, Republic of Korea), which is a member of the Korea Biobank Network.

Availability of data and materials

The data generated and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

JP and SL drafted the manuscript and revised it critically for important intellectual content. JP and SS performed the majority of the experiments and analyzed data. JH and HY performed the molecular experiments and interpreted data. SL and JK contributed to the conception and design of the work. JK gave the final approval of the version to be published. All authors agreed to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study protocol was approved by the Institutional Review Board of The Catholic University of Korea (DC18SESI0113). All subjects provided written informed consent for clinical and molecular analyses and publication before the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Liang H and Kim YH: Identifying molecular drivers of gastric cancer through next-generation sequencing. Cancer Lett. 340:241–246. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Tan P and Yeoh KG: Genetics and molecular pathogenesis of gastric adenocarcinoma. Gastroenterology. 149:1153–1162.e1153. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Berlth F, Bollschweiler E, Drebber U, Hoelscher AH and Moenig S: Pathohistological classification systems in gastric cancer: Diagnostic relevance and prognostic value. World J Gastroenterol. 20:5679–5684. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Cancer Genome Atlas Research Network, . Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 513:202–209. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Katona BW and Rustgi AK: Gastric Cancer Genomics: Advances and future directions. Cell Mol Gastroenterol Hepatol. 3:211–217. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Arkenau HT: Gastric cancer in the era of molecularly targeted agents: Current drug development strategies. J Cancer Res Clin Oncol. 135:855–866. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Paasinen-Sohns A, Koelzer VH, Frank A, Schafroth J, Gisler A, Sachs M, Graber A, Rothschild SI, Wicki A, Cathomas G and Mertz KD: Single-center experience with a targeted next generation sequencing assay for assessment of relevant somatic alterations in solid tumors. Neoplasia. 19:196–206. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Williams HL, Walsh K, Diamond A, Oniscu A and Deans ZC: Validation of the Oncomine™ focus panel for next-generation sequencing of clinical tumour samples. Virchows Arch. 473:489–503. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Lee A, Lee SH, Jung CK, Park G, Lee KY, Choi HJ, Min KO, Kim TJ, Lee EJ and Lee YS: Use of the Ion AmpliSeq cancer hotspot panel in clinical molecular pathology laboratories for analysis of solid tumours: With emphasis on validation with relevant single molecular pathology tests and the Oncomine Focus Assay. Pathol Res Pract. 214:713–719. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Sim NL, Kumar P, Hu J, Henikoff S, Schneider G and Ng PC: SIFT web server: Predicting effects of amino acid substitutions on proteins. Nucleic Acids Res. 40:W452–W457. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Adzhubei I, Jordan DM and Sunyaev SR: Predicting functional effect of human missense mutations using PolyPhen-2. Curr Protoc Hum Genet. 7(Unit7.20)2013.PubMed/NCBI

13 

Forbes SA, Beare D, Bindal N, Bamford S, Ward S, Cole CG, Jia M, Kok C, Boutselakis H, De T, et al: COSMIC: High-resolution cancer genetics using the catalogue of somatic mutations in cancer. Curr Protoc Hum Genet. 91:10.11.1–10.11.37. 2016. View Article : Google Scholar

14 

Matsusaka S, Kobunai T, Yamamoto N, Chin K, Ogura M, Tanaka G, Matsuoka K, Ishikawa Y, Mizunuma N and Yamaguchi T: Prognostic impact of KRAS mutant type and MET amplification in metastatic and recurrent gastric cancer patients treated with first-line S-1 plus cisplatin chemotherapy. Genes Cancer. 7:27–35. 2016.PubMed/NCBI

15 

Serra O, Galán M, Ginesta MM, Calvo M, Sala N and Salazar R: Comparison and applicability of molecular classifications for gastric cancer. Cancer Treat Rev. 77:29–34. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Alessandrini L, Manchi M, De Re V, Dolcetti R and Canzonieri V: Proposed molecular and miRNA classification of gastric cancer. Int J Mol Sci. 19:16832018. View Article : Google Scholar

17 

Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, Liu J, Yue YG, Wang J, Yu K, et al: Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 21:449–456. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Wang K, Yuen ST, Xu J, Lee SP, Yan HH, Shi ST, Siu HC, Deng S, Chu KM, Law S, et al: Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet. 46:573–582. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Salomao M, Luna AM, Sepulveda JL and Sepulveda AR: Mutational analysis by next generation sequencing of gastric type dysplasia occurring in hyperplastic polyps of the stomach: Mutations in gastric hyperplastic polyps. Exp Mol Pathol. 99:468–473. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Ge S, Li B, Li Y, Li Z, Liu Z, Chen Z, Wu J, Gao J and Shen L: Genomic alterations in advanced gastric cancer endoscopic biopsy samples using targeted next-generation sequencing. Am J Cancer Res. 7:1540–1553. 2017.PubMed/NCBI

21 

Ichikawa H, Nagahashi M, Shimada Y, Hanyu T, Ishikawa T, Kameyama H, Kobayashi T, Sakata J, Yabusaki H, Nakagawa S, et al: Actionable gene-based classification toward precision medicine in gastric cancer. Genome Med. 9:932017. View Article : Google Scholar : PubMed/NCBI

22 

Engelman JA, Luo J and Cantley LC: The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat Rev Genet. 7:606–619. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Shi J, Yao D, Liu W, Wang N, Lv H, Zhang G, Ji M, Xu L, He N, Shi B and Hou P: Highly frequent PIK3CA amplification is associated with poor prognosis in gastric cancer. BMC Cancer. 12:502012. View Article : Google Scholar : PubMed/NCBI

24 

Kim JW, Lee HS, Nam KH, Ahn S, Kim JW, Ahn SH, Park DJ, Kim HH and Lee KW: PIK3CA mutations are associated with increased tumor aggressiveness and Akt activation in gastric cancer. Oncotarget. 8:90948–90958. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Jang SH, Kim KJ, Oh MH, Lee JH, Lee HJ, Cho HD, Han SW, Son MW and Lee MS: Clinicopathological significance of elevated PIK3CA expression in gastric cancer. J Gastric Cancer. 16:85–92. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Thumkeo D, Watanabe S and Narumiya S: Physiological roles of Rho and Rho effectors in mammals. Eur J Cell Biol. 92:303–315. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Wang K, Kan J, Yuen ST, Shi ST, Chu KM, Law S, Chan TL, Kan Z, Chan AS, Tsui WY, et al: Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nat Genet. 43:1219–1223. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Deng N, Goh LK, Wang H, Das K, Tao J, Tan IB, Zhang S, Lee M, Wu J, Lim KH, 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. 61:673–684. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Pai P, Rachagani S, Dhawan P and Batra SK: Mucins and Wnt/β-catenin signaling in gastrointestinal cancers: An unholy nexus. Carcinogenesis. 37:223–232. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Li X, Wu WK, Xing R, Wong SH, Liu Y, Fang X, Zhang Y, Wang M, Wang J, Li L, et al: Distinct subtypes of gastric cancer defined by molecular characterization include novel mutational signatures with prognostic capability. Cancer Res. 76:1724–1732. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Horak P, Frohling S and Glimm H: Integrating next-generation sequencing into clinical oncology: Strategies, promises and pitfalls. ESMO Open. 1:e0000942016. View Article : Google Scholar : PubMed/NCBI

32 

Kuboki Y, Yamashita S, Niwa T, Ushijima T, Nagatsuma A, Kuwata T, Yoshino T, Doi T, Ochiai A and Ohtsu A: Comprehensive analyses using next-generation sequencing and immunohistochemistry enable precise treatment in advanced gastric cancer. Ann Oncol. 27:127–133. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Tirino G, Pompella L, Petrillo A, Laterza MM, Pappalardo A, Caterino M, Orditura M, Ciardiello F, Galizia G and De Vita F: What's new in gastric cancer: The therapeutic implications of molecular classifications and future perspectives. Int J Mol Sci. 19:26592018. View Article : Google Scholar

34 

Min L, Zhao Y, Zhu S, Qiu X, Cheng R, Xing J, Shao L, Guo S and Zhang S: Integrated analysis identifies molecular signatures and specific prognostic factors for different gastric cancer subtypes. Transl Oncol. 10:99–107. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Birkenkamp-Demtroder K, Hahn SA, Mansilla F, Thorsen K, Maghnouj A, Christensen R, Oster B and Orntoft TF: Keratin23 (KRT23) knockdown decreases proliferation and affects the DNA damage response of colon cancer cells. PLoS One. 8:e735932013. View Article : Google Scholar : PubMed/NCBI

36 

Pellino A, Riello E, Nappo F, Brignola S, Murgioni S, Djaballah SA, Lonardi S, Zagonel V, Rugge M, Loupakis F and Fassan M: Targeted therapies in metastatic gastric cancer: Current knowledge and future perspectives. World J Gastroenterol. 25:5773–5788. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Ho SWT and Tan P: Dissection of gastric cancer heterogeneity for precision oncology. Cancer Sci. 110:3405–3414. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2020
Volume 20 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Park J, Lee S, Shin S, Hong JH, Yoo HM and Kim JG: Genetic profiling of somatic alterations by Oncomine Focus Assay in Korean patients with advanced gastric cancer. Oncol Lett 20: 129, 2020
APA
Park, J., Lee, S., Shin, S., Hong, J.H., Yoo, H.M., & Kim, J.G. (2020). Genetic profiling of somatic alterations by Oncomine Focus Assay in Korean patients with advanced gastric cancer. Oncology Letters, 20, 129. https://doi.org/10.3892/ol.2020.11990
MLA
Park, J., Lee, S., Shin, S., Hong, J. H., Yoo, H. M., Kim, J. G."Genetic profiling of somatic alterations by Oncomine Focus Assay in Korean patients with advanced gastric cancer". Oncology Letters 20.5 (2020): 129.
Chicago
Park, J., Lee, S., Shin, S., Hong, J. H., Yoo, H. M., Kim, J. G."Genetic profiling of somatic alterations by Oncomine Focus Assay in Korean patients with advanced gastric cancer". Oncology Letters 20, no. 5 (2020): 129. https://doi.org/10.3892/ol.2020.11990