Retrospective Study Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Aug 15, 2022; 14(8): 1528-1539
Published online Aug 15, 2022. doi: 10.4251/wjgo.v14.i8.1528
Profiling of gene fusion involving targetable genes in Chinese gastric cancer
Zhen-Hua Liu, Department of Medical Oncology, Fujian Medical University, Fuzhou 350001, Fujian Province, China
Bo-Wen Zhu, Min Shi, Yu-Rong Qu, Hong-Ling Yuan, Wei Li, Dan-Dan Zhao, Bao-Ming Wang, Chun-Yang Wang, Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
Xun-Jun He, Department of Genetics and Genomic Medicine, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
Xun-Jun He, Zheng-Chuang Liu, Hou-Quan Tao, Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
Jie Ma, Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang Province, China
Zheng-Chuang Liu, Hou-Quan Tao, Department of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
Tong-Hui Ma, Department of Translational Medicine, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
ORCID number: Zhen-Hua Liu (0000-0002-1820-0292); Jie Ma (0000-0003-3755-996X); Tong-Hui Ma (0000-0002-5414-0522).
Author contributions: Liu ZH and Ma TH designed the study and reviewed the manuscript; Liu ZH, Zhu BW, Shi M analyzed the clinical and gene fusions data and wrote the manuscript; He XJ, Ma J, Liu ZC, and Tao HQ provided clinical advice; Yuan HL, Li W, Zhao DD, Wang BM, and Wang CY reviewed the manuscript and provided advice; All authors have read and approved the final manuscript.
Institutional review board statement: The study was reviewed and approved by the Fujian Provincial Hospital, Institutional Review Board (Approval No. K2022-03-101).
Informed consent statement: All study participants or their legal guardian provided informed written consent about personal and medical data collection prior to study enrolment.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Data sharing statement: No additional data are available.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Tong-Hui Ma, PhD, Director, Department of Translational Medicine, Genetron Health (Beijing) Technology, Co. Ltd., 1-2/F, Building 11, Zone 1, 8 Life Science Parkway, Changping District, Beijing 102200, China. tonghuima0818@sina.com
Received: March 30, 2022
Peer-review started: March 30, 2022
First decision: June 2, 2022
Revised: June 14, 2022
Accepted: July 19, 2022
Article in press: July 19, 2022
Published online: August 15, 2022

Abstract
BACKGROUND

Approximately half of all new cases of gastric cancer (GC) and related deaths occur in China. More than 80% of patients with GC are diagnosed at an advanced stage, which results in poor prognosis. Although HER2-directed therapy and immune checkpoint inhibitors have been somewhat successful, new drugs are still needed for the treatment of GC. Notably, several gene fusion-targeted drugs have been approved by the United States Food and Drug Administration for solid tumors, including GC, such as larotrectinib for NTRK fusion-positive cancers and zenocutuzumab for NRG1 fusion-positive cancers. However, gene fusions involving targetable genes have not been well characterized in Chinese patients with GC.

AIM

To identify the profile of fusions involving targetable genes in Chinese patients with GC using clinical specimens and determine the distribution of patients with gene fusion variants among the molecular subtypes of GC.

METHODS

We retrospectively analyzed gene fusion events in tumor tissue samples from 954 Chinese patients with GC. Clinicopathological characteristics were obtained from their medical records. Genetic alterations, such as single nucleotide variants, indels, amplifications, and gene fusions, were identified using a targeted sequencing panel containing 825 genes. Fusions were validated by fluorescence in situ hybridization (FISH) using break-apart probes. The microsatellite instability (MSI) status was evaluated using MSIsensor from the targeted sequencing panel data. Tumor mutational burden (TMB) was calculated using the total number of nonsynonymous mutations divided by the total genomic targeted region. Chi-square analysis was used to determine the enrichment of gene fusions associated with the molecular subtypes of GC.

RESULTS

We found that 1.68% (16/954) of patients harbored 20 fusion events involving targetable genes. RARA fusions (n = 5) were the most common, followed by FGFR2, BRAF, MET, FGFR3, RET, ALK, EGFR, NTRK2, and NRG1 fusions. Two of the RARA fusions, EML4-ALK (E6:E20) and EGFR-SEPTIN14 (E7:E10), have been identified in other tumors but not in GC. Surprisingly, 18 gene fusion events were previously not reported in any cancer types. Twelve of the eighteen novel gene fusions included complete exons encoding functional domains of targetable genes, such as the tyrosine kinase domain of receptor tyrosine kinases and the DNA- and ligand-binding domains of RARA. Consistent with the results of detection using the targeted sequencing fusion panel, the results of FISH (fluorescence in situ hybridization) confirmed the rearrangement of FGFR2 and BRAF in tumors from patients 04 and 09, respectively. Genetic analysis indicated that the fusion genes were significantly enriched in patients with ERBB2 amplification (P = 0.02); however, there were no significant differences between fusion-positive and fusion-negative patients in age, sex, MSI status, and TMB.

CONCLUSION

We characterized the landscape of fusions involving targetable genes in a Chinese GC cohort and found that 1.68% of patients with GC harbor potential targetable gene fusions, which were enriched in patients with ERBB2 amplification. Gene fusion detection may provide a potential treatment strategy for patients with GC with disease progression following standard therapy.

Key Words: Gene fusion, Targetable genes, Gastric cancer, Chinese population, ERBB2 amplification

Core Tip: The proportion of patients with gene fusions in Chinese patients with gastric cancer (GC) has not yet been characterized. In our analysis, we found that 1.68% of such patients harbor fusions involving targetable genes. Moreover, these fusion genes were enriched in patients with ERBB2 amplification. Our study indicates that gene fusion detection may provide a novel approach for GC therapy.



INTRODUCTION

Gastric cancer (GC) is the fifth most frequent cancer and the third leading cause of cancer deaths worldwide, with more than one million new cases and approximately 769000 deaths in 2020[1]. The overall survival rate of patients with early stage disease is around 90% after surgical resection[2]; however, more than 80% of patients with GC are diagnosed at an advanced stage in China, which limits the effectiveness of the treatment[3]. Although chemotherapy has improved the survival of advanced-stage patients with GC, the objective response rate remains less than 40%, and the median overall survival is less than 12 mo[4]. Nevertheless, new targeted therapies are capable of improving the objective response rate and overall survival of patients with GC expressing certain targets[5].

Approximately 13%-22% of GCs exhibit HER2 overexpression or amplification[6-8]. The College of American Pathologists, the American Society for Clinical Pathology, and the American Society of Clinical Oncology recommend that all patients with advanced gastric adenocarcinoma should be tested for HER2 overexpression[9]. Trastuzumab was approved by the United States Food and Drug Administration (FDA) in 2010 as first-line treatment in combination with chemotherapy for patients with HER2-positive GC. Microsatellite instability-high (MSI-H) tumors are considered a molecular subtype of gastric adenocarcinoma by The Cancer Genome Atlas (TCGA)[10]. The incidence of MSI-H GC is 10%-20%[11]. The NCCN guidelines recommend MSI testing as a standard test for all patients with GC. Regarding targeted therapy, the FDA has approved pembrolizumab (PD1 monoclonal antibody) for the treatment of all unresectable or metastatic solid tumors with MSI-H/dMMR (deficient DNA mismatch repair), including GC. Although drug treatments have shown success to some extent, the development of more targeted drugs is required.

With rapid advancements in the field of oncogenomics, gene fusions in cancer have received increasing attention. The FDA has approved larotrectinib (Vitrakvi) and entrectinib (Rozlytrek) for the first- or subsequent-line treatment of solid tumors with NTRK fusions, including GC[12,13]. In 2021, the FDA accelerated the approval of the NRG1 inhibitor, zenocutuzumab (MCLA-128), in patients with pan-cancer harboring an NRG1 fusion. Apart from these fusion genes with approved drugs in pan-cancer, ALK fusions, such as EML4-ALK, TFG-ALK, and STRN-ALK, have been identified in the majority of tumors, including lung adenocarcinoma and colorectal cancer[14-16]. For lung cancer and mesenchymal tumors, patients harboring an ALK fusion are highly responsive to crizotinib and ceritinib[17,18]. Recently, a RAB10-ALK fusion was identified in a patient with GC[19], which indicates the possibility of future applications of ALK-TKIs (tyrosine kinase inhibitors) in these patients. Recent advances in next-generation sequencing (NGS) have contributed to a surge in the discovery of fusion genes, including BRAF; EGFR; FGFR1, 2, and 3; RET; and ROS1[20]. Gene fusion detection can guide the development of targeted therapeutic strategies for patients with GC with disease progression after standard therapy. Notably, there is a lack of comprehensive data characterizing gene fusions involving targetable genes in GC, particularly in the Chinese population.

MATERIALS AND METHODS
Patients

This multicenter retrospective study included 1341 patients with GC admitted to Fujian Provincial Hospital (Fuzhou, China) and Zhejiang Provincial People’s Hospital (Hangzhou, China) between October 2015 and December 2021. The clinicopathological characteristics of the patients were retrieved from their medical records. Additionally, MSI status and tumor mutational burden (TMB) scores were extracted for statistical analysis. This study was approved by the Ethics Committee of the Fujian Provincial Hospital.

Mutational profiling

Mutational profiling of the Onco PanScan panel was performed by Genetron Health (Beijing) Co., Ltd. The coding regions of 825 cancer-related genes were analyzed. Genomic DNA was isolated from formalin-fixed paraffin-embedded (FFPE) tissue specimens with a minimum of 20% viable tumor nuclei. For sequencing, paired tumor and white blood cell DNA libraries were prepared using KAPA HyperPrep Kits (Roche, Germany). Libraries were quantified using Qubit fluorometer (Thermo Fisher Scientific, Waltham, MA, United States), and their quality was evaluated using Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, United States). High-throughput sequencing was performed on Novaseq6000 platform (Illumina, United States). Paired-end reads from Illumina sequencing were processed using script bcl2fastq (v. 2.17.1.14) and aligned against the human genome reference build, GRCh37, using Burrows-Wheeler Aligner (BWA, version 0.7.13). Duplicate removal, local realignment, and base quality recalibration were performed using PICARD (http://broadinstitute.github.io/picard/) and the Genome Analysis Toolkit. Variant calling was performed using an in-house developed pipeline. Variants identified as germline variants were excluded, while single nucleotide variants (SNVs) and indels with allelic fractions of more than 5% and supported by more than 4 unique reads, amplification with a fold-change greater than 2.5 in more than 25% of regions covered, and gene fusions supported by more than 3 unique reads were included.

TMB was calculated using the total number of nonsynonymous mutations divided by the total genomic target region (2.13 Mb). MSI status was determined using MSIsensor from paired tumor-normal targeted sequence data, and 309 MSI sites were included in the panel of 825 cancer-related genes. An MSIsensor score below 10 defines microsatellite stability (MSS) status, while that above 50 defines MSI status. The prevalence of gene fusions involving a targetable gene and driver mutations was compared with the OrigiMed2020 and TCGA cohorts[21]. Clinicopathological and genomic data were retrieved from the cBioPortal (https://www.cbioportal.org).

Fluorescence in situ hybridization

FFPE tissue sections (5 μm) were prepared on positively charged slides. After deparaffinizing and rehydrating, the slides were incubated with prewarmed 8% sodium thiocyanate in dH2O at 80 °C and incubated for 30 min. FGFR2 (10q26) or BRAF (7q34) break-apart probes were placed on the slide, covered with a glass coverslip, and sealed with rubber cement. Hybridization was performed overnight at 37 °C. The slides were washed twice in 50% formamide at 47 °C for 2 min and then twice in 2X standard saline citrate at room temperature for 2 min. Nuclei were stained with DAPI as a counterstain. The slides were scanned using a 90i Nikon fluorescent microscope. For each probe, 200 nuclei were evaluated. The 5′ (red) and 3′ (green) signals separated by ≥ 2 signal diameters were considered split as positive.

Statistical analysis

All statistical analyses were performed using SPSS 24.0 software (IBM, Chicago, IL, United States). χ2 or Fisher’s exact test was used to analyze the association between fusion alterations and driver mutations. A P value of < 0.05 was considered statistically significant.

RESULTS
Clinical characteristics of patients

We retrospectively analyzed 1341 Chinese patients with GC who underwent genetic analysis from multiple centers in China. Of these, 387 patients were excluded because gene fusion detection was not performed with the Onco PanScan panel using tumor tissue samples (Figure 1). Gene fusion events were detected in 20 patients; however, 4 patients without any gene fusions involving targetable genes were excluded. Finally, 16 patients with 20 fusion events involving targetable genes were included for further analysis. The clinical characteristics of 954 patients with GC are shown in Table 1. Of these patients, 310 (32.56%) were women and 644 (67.44%) were men, with a median age of 57 and 62, respectively, at diagnosis. There was no significant difference between targetable gene fusion-positive and -negative patients in age (P = 0.293), sex (P = 0.463), MSI status (P = 0.551), or TMB (P = 0.217) (Table 1).

Figure 1
Figure 1 Flowchart of patient selection. GC: Gastric cancer.
Table 1 Clinical characteristics in targetable gene fusion-positive and -negative patients.
Variables
Total, n
Fusion involving targetable genes
P value
Positive, n (%)
Negative, n (%)
Sex0.293
Female3103 (0.97)307 (99.03)
Male64413 (2.02)631 (97.98)
Age, yr0.463
≤ 604516 (1.56)445 (98.44)
> 6050310 (1.98)493 (98.01)
MSI status0.551
MSI-H461 (2.17)45 (97.93)
MSS90815 (1.65)893 (98.35)
TMB0.217
Median TMB score2.925.632.83
The landscape of gene fusions involving targetable genes in Chinese patients with GC

To gain insight into fusion events in GC, we evaluated 954 patients with GC undergoing gene fusion analysis. In total, 20 patients harbored 24 gene fusions, 2 patients had double fusions (patient 01 and 02), and 1 patient (09) harbored triple fusions. RARA fusions (5/24, 17.8%) and FGFR family gene fusions (5/24, 17.8%) occurred most frequently in the cohort, followed by BRAF (3/24, 10.7%) and MET (2/24, 8.3%) (Figure 2A). ALK, RET, NTRK2, NRG1, and EGFR fusions were identified in one patient each. Remarkably, 20 of 24 (83.3%) fusions involved targetable genes (Table 2). RARA has been frequently reported as a 3′ fusion partner in acute promyelocytic leukemia[22]. RARA was identified as a 5′ fusion partner in 4 patients and as a 3′ fusion partner in 1 patient; however, only the KRPAT9-RARA fusion was detected in patient 01 as the 3′ fusion partner including exons 3-9, which encodes a DNA-binding and a ligand-binding domain required for RARA transcription factor activity[22]. Three BRAF fusions were identified in patient 09 as the 3′ fusion partner containing the complete tyrosine kinase domain, which was coded by exons 11-18. All FGFR2 and FGFR3 fusions were detected as 5′ fusion partners. Four FGFR2 fusions were consistent with other known activating FGFR2 fusions[23], which frequently occur with a breakpoint after exon 17 at the 3′ end of FGFR2 with a 3′ fusion partner. The kinase domain was retained in these fusion genes. In patient 10, the MET fusion involved the 5′ end of MET exon 7, thus retaining an intact MET kinase domain.

Figure 2
Figure 2 Profile of targetable gene fusions in gastric cancer. A: The types and proportion of 24 gene fusions. Others included targetable ALK, RET, NTRK2, NRG1, and EGFR fusions. Four fusions without targetable genes were excluded from the analysis; B: Comparison of gene fusion frequencies in our cohort and the OrigiMed2020 and The Cancer Genome Atlas (TCGA) cohorts. No statistical differences were found among the cohorts.
Table 2 List of gene fusions involving targetable genes in Chinese patients with gastric cancer and drugs under clinical trial or approved by the Food and Drug Administration.
Patients ID
Fusion gene
5' partner gene
3' partner gene
Variant frequency, %
Functional domain is included or not
Targeted drugs
Gene name
Chromosome
Last observed exon
Breakpoint
Gene name
Chromosome
First observed exon
Breakpoint
Patient 01RARA-PGAP3RARA17338504951PGAP31783782802024.1Partially includeTamibarotene targeting RARA fusion2
Patient 01KRTAP9-7-RARAKRTAP9-717downstream39437039RARA1733849954756.9Completely includeTamibarotene targeting RARA fusion2
Patient 02RARA-KRT13RARA17238491648KRT131783965726929.2Partially includeTamibarotene targeting RARA fusion2
Patient 02RARA-ETV4RARA17238499726ETV41754162124376Partially includeTamibarotene targeting RARA fusion2
Patient 03RARA-IKZF3RARA17238504120IKZF31723800955518.4Partially includeTamibarotene targeting RARA fusion2
Patient 04FGFR2-PDE2AFGFR21017123241248PDE2A117723072511.4Completely includePemigatinib; Erdafitinib targeting FGFR fusion1
Patient 05FGFR2-intergenicFGFR21017123242196intergenic10-12339410716.6Completely includePemigatinib; Erdafitinib targeting FGFR fusion1
Patient 06FGFR2-intergenicFGFR21017123240841intergenic10-1227938424.2Completely includePemigatinib; Erdafitinib targeting FGFR fusion1
Patient 07FGFR2-SHTN1FGFR21017123242528SHTN11061187093055.1Completely includePemigatinib; Erdafitinib targeting FGFR fusion1
Patient 08FGFR3-PHTF2FGFR34181808927PHTF2711775679823.3Completely includePemigatinib; Erdafitinib targeting FGFR fusion1
Patient 09STIM2-BRAFSTIM241127012641BRAF7914048792912.7Completely includeSelumetinib targeting BRAF fusion1
Patient 09STIM2-BRAFSTIM241127013243BRAF7101404861031.1Completely includeSelumetinib targeting BRAF fusion1
Patient 09TBC1D19-BRAFTBC1D194426629603BRAF7101404867826.5Completely includeSelumetinib targeting BRAF fusion1
Patient 10TES-METTES71115867013MET721163322270.7Completely includeCrizotinib targeting MET fusion1
Patient 11MET-TESMET721116436166TES7411588944524.6Not includeCrizotinib targeting MET fusion1
Patient 12EML4-ALKEML42629447382ALK220424986623.5Completely includeCrizotinib; ceritinib targeting ALK fusion1
Patient 13OPALIN-RETOPALIN10698104545RET1011436100995.46Completely includePralsetinib targeting RET fusion1
Patient 14ARHGAP10-NTRK2ARHGAP1041148716754NTRK29168747664515.3Completely includeLarotrectinib targeting NTRK2 fusion1
Patient 15NRG1-FDFT1NRG181232617907FDFT188116853758.9Partially includeMCLA-128 targeting NRG1 fusion2
Patient 16EGFR-SEPTIN14EGFR72555269173SEPTIN14710558711799.6Completely includeAfatinib targeting EGFR fusion2

The frequency of fusion events involving the abovementioned 10 targetable genes in the TCGA GC cohort and another Chinese GC cohort (OrigiMed2020 cohort) were analyzed and compared with our patient data (Figure 2B). Neither our cohort nor the OrigiMed2020 cohort showed significant differences in the incidence of these gene fusions in Chinese patients. In two Chinese cohorts, EGFR fusions occurred less frequently. Fusions in MET, BRAF, RET, ALK, and NTRK2 were only identified in two Chinese cohorts; however, the differences in the incidence of these genes were not statistically significant.

Novel fusions involving targetable genes in GC

In total, 2 of 20 fusions involving the targetable genes, EML4-ALK and EGFR-SEPTIN14, were reported in other cancers, including non-small-cell lung cancer[24-26]. The remaining 18 gene fusions were not reported in any cancer types. In total, 13 of 18 novel gene fusions contained the exon encoding a tyrosine kinase domain, such as exons 11-17 of FGFR2, exons 11-18 of BRAF, exons 12-19 of MET, and exons 16-21 of NTRK2 (Figure 3). All fusions involving FGFR2, BRAF, RET, and NTRK2 retained the kinase domain (Table 2). Furthermore, reads in Integrative Genomics Viewer plots supported these gene fusions. To verify these novel fusions, fluorescence in situ hybridization (FISH) was performed using break-apart probes. Because only two tumor tissue samples were available, only the FGFR2 and BRAF arrangement in patient 04 and 09, respectively, were confirmed by FISH.

Figure 3
Figure 3 Examples of novel gene fusions involving targetable genes in gastric cancer. A-D: Schematic representation and Integrative Genomics Viewer screenshot of FGFR2-PED2A (A), STIM-BRAF (B), OPALIN-RET (C), and NTRK2-ARHGAP10 (D) are shown; A and B: FGFR2 and BRAF fusions were confirmed by fluorescence in situ hybridization using FGFR2 (10q26) or BRAF (7q34) break-apart probes. Red spot: 5′ Probe signal; Green spot: 3′ probe signal; Yellow spot: Target gene without rearrangement. Arrows indicate the cells with separate 5′ (red) and 3′ (green) signals. Bar: 100 μm. TK: Tyrosine kinase domain.
Gene fusions are enriched in patients with ERBB2 amplification but not in those with high MSI and TMB

Because of the low frequency of gene fusions in patients with GC, we determined whether gene fusions are enriched in different molecular subtypes of GC, which may indicate the patients that could benefit from gene fusion detection. Fusions are mutually exclusive with other oncogenic mutations and are enriched in patients without driver mutations[27-29]. In our cohort, the frequency of genetic alterations in oncogenic driver genes of GC, such as TP53, ARID1A, CDH1, and PIK3CA mutations and ERBB2 amplification, were comparable with those in the TCGA cohort (Supplementary Figure 1). There was no significant difference in the frequency of fusions involving targetable genes between patients with any alterations in all five driver genes and those without (Figure 4A). Notably, the fusion alteration frequency was significantly higher in patients with ERBB2 amplification than in those without ERBB2 amplification (Figure 4B, P = 0.01). To determine whether fusion alterations were enriched in other driver genes, TP53, ARID1A, CDH1, and PIK3CA were analyzed. There was no enrichment in fusion alterations for these genes (Supplementary Figure 2). Forty-six patients had the MSI-H phenotype. Of these, one patient with fusion genes exhibited MSI-H. There was no obvious difference in the incidence of gene fusions between patients with MSI-H and MSS (Figure 4C). Similarly, TMB scores were evaluated in targetable gene fusion-positive and -negative patients, but the results were not statistically significant (Figure 4D).

Figure 4
Figure 4 Enrichment of gene fusions in patients with gastric cancer with driver alterations. A: The incidence of gene fusions in patients with and without driver alterations were analyzed, P > 0.05; B: The incidence of gene fusions in patients with and without ERBB2 amplifications were analyzed, P < 0.05; C: The incidence of gene fusions in patients with microsatellite instability-high and microsatellite stability were analyzed, P > 0.05; D: Tumor mutational burden in targetable gene fusion-positive and -negative patients was compared, P > 0.05.
DISCUSSION

Structural gene rearrangements leading to gene fusions are common events that occur in solid tumors. Gene fusions have been considered oncogenic drivers in neoplasia for more than 30 years[30]. Detection and characterization of gene fusions is important for clinical purposes[31]. As the first large-scale study focusing on gene fusion events in Chinese patients with GC, we retrospectively analyzed 954 tumor specimens to identify fusions involving targetable genes and confirmed the occurrence of these fusions in GC.

In this study, 16 of 954 patients harbored 20 fusions involving targetable genes, the majority of which had not been previously reported, including FGFR2-PDE2A, STIM2-BRAF, OPALIN-RET, and ARHGAP10-NTRK2. However, we did not find any significant differences between the Chinese GC cohort (our cohort and OrigiMed2020 cohort) and the TCGA cohort. Fusions in BRAF, RET, ALK, and NTRK2 were detected in two Chinese cohorts but not in the TCGA cohort. This finding may have resulted from the small size of the TCGA cohort, which is prone to bias for gene fusions events because of the low occurrence rate in GC. A comparative study with a larger population is needed to identify differences in fusions involving targetable genes between races.

A major contribution of gene fusions to patients with tumor is the development of drugs that target fusion proteins encoded by these genes. The majority of advances in targeting gene fusions involve kinase domains that constitutively activate downstream signaling pathways[32]. In this study, except RARA and NRG1 fusions, the 14 other fusions involving targetable genes included a receptor tyrosine kinase (RTK) gene, such as FGFR2/3, BRAF, MET, ALK, RET, NTRK2, and EGFR. Furthermore, most of all RTK gene fusions (13/14) completely retained the tyrosine kinase domain, which resulted in functional fusion proteins. We only verified the BRAF rearrangement in patient 09 and the FGFR2 rearrangement in patient 04 using FISH because of insufficient tumor specimens. These fusions were consistent with previously observed fusions[23]; however, only 1 out of 5 RARA fusions contained exons 3-9, which encodes a DNA-binding and ligand-binding domain, which are required for RARA transcription factor activity. These results indicate that most patients with GC with fusions involving targetable genes may benefit from drugs that target fusions. However, patients in this retrospective study had not received targeted drug treatment; thus, we cannot determine whether they would have benefited from fusion-targeted drug therapy.

Interestingly, we also discovered 18 novel fusions with unreported partner genes or with an intergenic space. In other words, screening for known fusions in GC by FISH or polymerase chain reaction will likely miss most of the gene fusions that involve targetable genes. This is not conducive to patients with GC participating in clinical trials of fusion-targeted drugs in pan-cancer. Additionally, we found gene fusions enriched in patients with ERBB2 amplification. We did not confirm all fusions using FISH because of limited tumor tissue samples, nor could we identify gene fusions enriched in distinct molecular subtypes of GC. Moreover, the efficacy of fusion-targeted drugs in GC remains to be further validated in clinical trials. Despite these limitations, for patients who fail standard therapy, NGS-based novel gene fusion detection may provide a new treatment strategy and facilitate participation into clinical trials involving targeted therapy.

CONCLUSION

As the first large-scale study focusing on gene fusion events in Chinese patients with GC, we determined the frequency (16/954) of targetable gene fusions, and the majority of these fusions, including TES-MET, FGFR2-PDE2A, OPALIN-RET, STIM-BRAF, ARHGAP10-NTRK2, and EGFR-SEPTIN14, had not been previously described. These novel fusions completely retain a kinase domain. Additionally, we found gene fusions that were enriched in patients with ERBB2 amplification. Gene fusion detection may aid in the development of novel treatment strategies for patients with GC.

ARTICLE HIGHLIGHTS
Research background

With rapid advancements in oncogenomics, increasing attention has been focused on gene fusions in cancer. The Food and Drug Administration has approved several fusion-targeted drugs for the treatment of solid tumors, such as larotrectinib for NTRK fusion-positive cancers and Zenocutuzumab for NRG1 fusion-positive cancers. However, targetable gene fusions in Chinese patients with gastric cancer (GC) have not been well characterized.

Research motivation

To investigate the incidence of gene fusions involving targetable genes in Chinese patients with GC and explore a potential treatment strategy for patients with GC.

Research objectives

To explore the types and proportion of targetable gene fusions in Chinese patients with GC and determine the distribution of patients with gene fusions among the molecular subtypes of GC.

Research methods

This was a multicenter retrospective study that evaluated patients with GC. A total of 954 tumor tissue samples from patients with GC who underwent gene fusion detection were included. Genetic alterations, including SNVs, indels, amplifications, and gene fusions, were analyzed. The enrichment of gene fusions in the molecular subtypes of GC was explored.

Research results

Twenty fusions involving targetable genes were detected. Among them, 18 novel gene fusion events were previously not reported in other cancers. Owing to a limited number of tumor tissue samples, only BRAF and FGFR2 fusions were identified by fluorescence in situ hybridization. Additionally, we found that gene fusions were enriched in patients with ERBB2 amplification.

Research conclusions

Gene fusions involving targetable genes were characterized in Chinese patients with GC. Testing gene fusions may provide insight for the treatment of GC.

Research perspectives

A large study should be performed to further confirm the targetable gene fusions and identify whether gene fusions are enriched in distinct molecular subtypes of GC.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Corresponding Author's Membership in Professional Societies: American Association for Cancer Research, No. 1070947.

Specialty type: Oncology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Goebel WS, United States; Park J, South Korea S-Editor: Gong ZM L-Editor: Filipodia P-Editor: Gong ZM

References
1.  Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50630]  [Cited by in F6Publishing: 43243]  [Article Influence: 14414.3]  [Reference Citation Analysis (47)]
2.  Sexton RE, Al Hallak MN, Diab M, Azmi AS. Gastric cancer: a comprehensive review of current and future treatment strategies. Cancer Metastasis Rev. 2020;39:1179-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in F6Publishing: 277]  [Article Influence: 69.3]  [Reference Citation Analysis (0)]
3.  Jin G, Lv J, Yang M, Wang M, Zhu M, Wang T, Yan C, Yu C, Ding Y, Li G, Ren C, Ni J, Zhang R, Guo Y, Bian Z, Zheng Y, Zhang N, Jiang Y, Chen J, Wang Y, Xu D, Zheng H, Yang L, Chen Y, Walters R, Millwood IY, Dai J, Ma H, Chen K, Chen Z, Hu Z, Wei Q, Shen H, Li L. Genetic risk, incident gastric cancer, and healthy lifestyle: a meta-analysis of genome-wide association studies and prospective cohort study. Lancet Oncol. 2020;21:1378-1386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 95]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
4.  Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23762]  [Cited by in F6Publishing: 25182]  [Article Influence: 1937.1]  [Reference Citation Analysis (3)]
5.  Patel TH, Cecchini M. Targeted Therapies in Advanced Gastric Cancer. Curr Treat Options Oncol. 2020;21:70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 110]  [Article Influence: 27.5]  [Reference Citation Analysis (0)]
6.  Grillo F, Fassan M, Sarocchi F, Fiocca R, Mastracci L. HER2 heterogeneity in gastric/gastroesophageal cancers: From benchside to practice. World J Gastroenterol. 2016;22:5879-5887.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 76]  [Cited by in F6Publishing: 74]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
7.  Abrahao-Machado LF, Scapulatempo-Neto C. HER2 testing in gastric cancer: An update. World J Gastroenterol. 2016;22:4619-4625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 155]  [Cited by in F6Publishing: 162]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
8.  Rüschoff J, Hanna W, Bilous M, Hofmann M, Osamura RY, Penault-Llorca F, van de Vijver M, Viale G. HER2 testing in gastric cancer: a practical approach. Mod Pathol. 2012;25:637-650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 379]  [Cited by in F6Publishing: 394]  [Article Influence: 32.8]  [Reference Citation Analysis (0)]
9.  Bartley AN, Washington MK, Colasacco C, Ventura CB, Ismaila N, Benson AB 3rd, Carrato A, Gulley ML, Jain D, Kakar S, Mackay HJ, Streutker C, Tang L, Troxell M, Ajani JA. HER2 Testing and Clinical Decision Making in Gastroesophageal Adenocarcinoma: Guideline From the College of American Pathologists, American Society for Clinical Pathology, and the American Society of Clinical Oncology. J Clin Oncol. 2017;35:446-464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in F6Publishing: 232]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
10.  Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4230]  [Cited by in F6Publishing: 4299]  [Article Influence: 429.9]  [Reference Citation Analysis (2)]
11.  Cho J, Kang SY, Kim KM. MMR protein immunohistochemistry and microsatellite instability in gastric cancers. Pathology. 2019;51:110-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 20]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
12.  Scott LJ. Larotrectinib: First Global Approval. Drugs. 2019;79:201-206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 141]  [Article Influence: 28.2]  [Reference Citation Analysis (0)]
13.  Frampton JE. Entrectinib: A Review in NTRK+ Solid Tumours and ROS1+ NSCLC. Drugs. 2021;81:697-708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
14.  Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S, Watanabe H, Kurashina K, Hatanaka H, Bando M, Ohno S, Ishikawa Y, Aburatani H, Niki T, Sohara Y, Sugiyama Y, Mano H. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448:561-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3816]  [Cited by in F6Publishing: 3912]  [Article Influence: 230.1]  [Reference Citation Analysis (0)]
15.  Roskoski R Jr. Anaplastic lymphoma kinase (ALK): structure, oncogenic activation, and pharmacological inhibition. Pharmacol Res. 2013;68:68-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 198]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
16.  Holla VR, Elamin YY, Bailey AM, Johnson AM, Litzenburger BC, Khotskaya YB, Sanchez NS, Zeng J, Shufean MA, Shaw KR, Mendelsohn J, Mills GB, Meric-Bernstam F, Simon GR. ALK: a tyrosine kinase target for cancer therapy. Cold Spring Harb Mol Case Stud. 2017;3:a001115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 109]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
17.  Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, Ou SH, Dezube BJ, Jänne PA, Costa DB, Varella-Garcia M, Kim WH, Lynch TJ, Fidias P, Stubbs H, Engelman JA, Sequist LV, Tan W, Gandhi L, Mino-Kenudson M, Wei GC, Shreeve SM, Ratain MJ, Settleman J, Christensen JG, Haber DA, Wilner K, Salgia R, Shapiro GI, Clark JW, Iafrate AJ. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010;363:1693-1703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3425]  [Cited by in F6Publishing: 3460]  [Article Influence: 247.1]  [Reference Citation Analysis (0)]
18.  Camidge DR, Bang YJ, Kwak EL, Iafrate AJ, Varella-Garcia M, Fox SB, Riely GJ, Solomon B, Ou SH, Kim DW, Salgia R, Fidias P, Engelman JA, Gandhi L, Jänne PA, Costa DB, Shapiro GI, Lorusso P, Ruffner K, Stephenson P, Tang Y, Wilner K, Clark JW, Shaw AT. Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: updated results from a phase 1 study. Lancet Oncol. 2012;13:1011-1019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 923]  [Cited by in F6Publishing: 977]  [Article Influence: 81.4]  [Reference Citation Analysis (0)]
19.  Wen Z, Xiong D, Zhang S, Liu J, Li B, Li R, Zhang H. Case Report: RAB10-ALK: A Novel ALK Fusion in a Patient With Gastric Cancer. Front Oncol. 2021;11:645370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
20.  Stransky N, Cerami E, Schalm S, Kim JL, Lengauer C. The landscape of kinase fusions in cancer. Nat Commun. 2014;5:4846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 605]  [Cited by in F6Publishing: 680]  [Article Influence: 68.0]  [Reference Citation Analysis (0)]
21.  Hoadley KA, Yau C, Hinoue T, Wolf DM, Lazar AJ, Drill E, Shen R, Taylor AM, Cherniack AD, Thorsson V, Akbani R, Bowlby R, Wong CK, Wiznerowicz M, Sanchez-Vega F, Robertson AG, Schneider BG, Lawrence MS, Noushmehr H, Malta TM; Cancer Genome Atlas Network, Stuart JM, Benz CC, Laird PW. Cell-of-Origin Patterns Dominate the Molecular Classification of 10,000 Tumors from 33 Types of Cancer. Cell. 2018;173:291-304.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1450]  [Cited by in F6Publishing: 1308]  [Article Influence: 218.0]  [Reference Citation Analysis (0)]
22.  De Braekeleer E, Douet-Guilbert N, De Braekeleer M. RARA fusion genes in acute promyelocytic leukemia: a review. Expert Rev Hematol. 2014;7:347-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 67]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
23.  Fusco MJ, Saeed-Vafa D, Carballido EM, Boyle TA, Malafa M, Blue KL, Teer JK, Walko CM, McLeod HL, Hicks JK, Extermann M, Fleming JB, Knepper TC, Kim DW. Identification of Targetable Gene Fusions and Structural Rearrangements to Foster Precision Medicine in KRAS Wild-Type Pancreatic Cancer. JCO Precis Oncol. 2021;5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
24.  Koivunen JP, Mermel C, Zejnullahu K, Murphy C, Lifshits E, Holmes AJ, Choi HG, Kim J, Chiang D, Thomas R, Lee J, Richards WG, Sugarbaker DJ, Ducko C, Lindeman N, Marcoux JP, Engelman JA, Gray NS, Lee C, Meyerson M, Jänne PA. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin Cancer Res. 2008;14:4275-4283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 729]  [Cited by in F6Publishing: 785]  [Article Influence: 49.1]  [Reference Citation Analysis (0)]
25.  Cantile M, Marra L, Franco R, Ascierto P, Liguori G, De Chiara A, Botti G. Molecular detection and targeting of EWSR1 fusion transcripts in soft tissue tumors. Med Oncol. 2013;30:412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 58]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
26.  Zhu YC, Wang WX, Li XL, Xu CW, Chen G, Zhuang W, Lv T, Song Y. Identification of a Novel Icotinib-Sensitive EGFR-SEPTIN14 Fusion Variant in Lung Adenocarcinoma by Next-Generation Sequencing. J Thorac Oncol. 2019;14:e181-e183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
27.  Chou A, Fraser T, Ahadi M, Fuchs T, Sioson L, Clarkson A, Sheen A, Singh N, Corless CL, Gill AJ. NTRK gene rearrangements are highly enriched in MLH1/PMS2 deficient, BRAF wild-type colorectal carcinomas-a study of 4569 cases. Mod Pathol. 2020;33:924-932.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
28.  Wang Y, Xu Y, Wang X, Sun C, Guo Y, Shao G, Yang Z, Qiu S, Ma K. RET fusion in advanced non-small-cell lung cancer and response to cabozantinib: A case report. Medicine (Baltimore). 2019;98:e14120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
29.  Kobayashi M, Sakakibara T, Inoue A, Fukuhara T, Sasano H, Ichinose M, Nukiwa T. Effective enrichment strategy for EML4-ALK fusion gene screening in patients with non-small cell lung cancer. Respir Investig. 2014;52:49-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
30.  Mertens F, Johansson B, Fioretos T, Mitelman F. The emerging complexity of gene fusions in cancer. Nat Rev Cancer. 2015;15:371-381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 439]  [Cited by in F6Publishing: 437]  [Article Influence: 48.6]  [Reference Citation Analysis (0)]
31.  Mitelman F, Johansson B, Mertens F. The impact of translocations and gene fusions on cancer causation. Nat Rev Cancer. 2007;7:233-245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1017]  [Cited by in F6Publishing: 950]  [Article Influence: 55.9]  [Reference Citation Analysis (0)]
32.  Schram AM, Chang MT, Jonsson P, Drilon A. Fusions in solid tumours: diagnostic strategies, targeted therapy, and acquired resistance. Nat Rev Clin Oncol. 2017;14:735-748.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 208]  [Article Influence: 29.7]  [Reference Citation Analysis (0)]