Research article

Family with sequence similarity 153 member B as a potential prognostic biomarker of gastric cancer


  • Received: 06 July 2022 Revised: 12 August 2022 Accepted: 18 August 2022 Published: 29 August 2022
  • Gastric cancer (GC) is one of the most common digestive tumors in Northwest China. Previous sequencing analysis revealed that family with sequence similarity 153 member B (FAM153B) might be the primary driver gene of GC. In this study, we aim to explore the potential roles of FAM153B in GC. Microarray data were firstly obtained from public databases with the aim to evaluate the genetic expression of FAM153B between GC and normal tissues. The results were verified in immunohistochemistry (IHC). We also performed the co-expression network analysis and enrichment analysis to identify underlying mechanisms. A correlation analysis of FAM153B expression and immune infiltration was performed then. Furthermore, two GC cell lines were used to evaluate the effect of FAM153B on gastric cell proliferation by employing MTT and Edu assays. Our findings suggest that FAM153B is downregulated in tumoral tissue, and positively associated with unfavorable survival. The enrichment pathways of FAM153B were regulation of signaling receptor activity, DNA replication, cell cycle transition, chromosomal regulation, and so on. Besides, from the perspective of bioinformatics, the protein expression level of FAM153B is related to the degree of immune cell infiltration. In vitro, overexpression of FAM153B inhibit the proliferation of two cell lines. In summary, this study demonstrates that FAM153B might serve as an effective prognostic and therapeutic biomarker in GC.

    Citation: Yuan Yang, Yuwei Ye, Min Liu, Ya Zheng, Guozhi Wu, Zhaofeng Chen, Yuping Wang, Qinghong Guo, Rui Ji, Yongning Zhou. Family with sequence similarity 153 member B as a potential prognostic biomarker of gastric cancer[J]. Mathematical Biosciences and Engineering, 2022, 19(12): 12581-12600. doi: 10.3934/mbe.2022587

    Related Papers:

    [1] Yuan Yang, Lingshan Zhou, Xi Gou, Guozhi Wu, Ya Zheng, Min Liu, Zhaofeng Chen, Yuping Wang, Rui Ji, Qinghong Guo, Yongning Zhou . Comprehensive analysis to identify DNA damage response-related lncRNA pairs as a prognostic and therapeutic biomarker in gastric cancer. Mathematical Biosciences and Engineering, 2022, 19(1): 595-611. doi: 10.3934/mbe.2022026
    [2] Kaiyu Shen, Shuaiyi Ke, Binyu Chen, Tiantian Zhang, Hongtai Wang, Jianhui Lv, Wencang Gao . Identification and validation of biomarkers for epithelial-mesenchymal transition-related cells to estimate the prognosis and immune microenvironment in primary gastric cancer by the integrated analysis of single-cell and bulk RNA sequencing data. Mathematical Biosciences and Engineering, 2023, 20(8): 13798-13823. doi: 10.3934/mbe.2023614
    [3] Guoxin Hou, Binbin Song . Gastric cancer patient with c-MET amplification treated with crizotinib after failed multi-line treatment: A case report and literature review. Mathematical Biosciences and Engineering, 2019, 16(5): 5923-5930. doi: 10.3934/mbe.2019296
    [4] Yi Liu, Long Cheng, Xiangyang Song, Chao Li, Jiantao Zhang, Lei Wang . A TP53-associated immune prognostic signature for the prediction of the overall survival and therapeutic responses in pancreatic cancer. Mathematical Biosciences and Engineering, 2022, 19(1): 191-208. doi: 10.3934/mbe.2022010
    [5] Xin Lin, Xingyuan Li, Binqiang Ma, Lihua Hang . Identification of novel immunomodulators in lung squamous cell carcinoma based on transcriptomic data. Mathematical Biosciences and Engineering, 2022, 19(2): 1843-1860. doi: 10.3934/mbe.2022086
    [6] Li Peng, Yujie Yang, Cheng Yang, Zejun Li, Ngai Cheong . HRGCNLDA: Forecasting of lncRNA-disease association based on hierarchical refinement graph convolutional neural network. Mathematical Biosciences and Engineering, 2024, 21(4): 4814-4834. doi: 10.3934/mbe.2024212
    [7] Jinsong Liu, Yuyang Dai, Yueyao Lu, Xiuling Liu, Jianzhong Deng, Wenbin Lu, Qian Liu . Identification and validation of a new pyroptosis-associated lncRNA signature to predict survival outcomes, immunological responses and drug sensitivity in patients with gastric cancer. Mathematical Biosciences and Engineering, 2023, 20(2): 1856-1881. doi: 10.3934/mbe.2023085
    [8] Ye Hu, Meiling Wang, Kainan Wang, Jiyue Gao, Jiaci Tong, Zuowei Zhao, Man Li . A potential role for metastasis-associated in colon cancer 1 (MACC1) as a pan-cancer prognostic and immunological biomarker. Mathematical Biosciences and Engineering, 2021, 18(6): 8331-8353. doi: 10.3934/mbe.2021413
    [9] Li-Sha Pan, Zacharia Ackbarkha, Jing Zeng, Min-Li Huang, Zhen Yang, Hao Liang . Immune marker signature helps to predict survival in uveal melanoma. Mathematical Biosciences and Engineering, 2021, 18(4): 4055-4070. doi: 10.3934/mbe.2021203
    [10] Jianpei Hu, Zengnan Mo . Dissection of tumor antigens and immune landscape in clear cell renal cell carcinoma: Preconditions for development and precision medicine of mRNA vaccine. Mathematical Biosciences and Engineering, 2023, 20(2): 2157-2182. doi: 10.3934/mbe.2023100
  • Gastric cancer (GC) is one of the most common digestive tumors in Northwest China. Previous sequencing analysis revealed that family with sequence similarity 153 member B (FAM153B) might be the primary driver gene of GC. In this study, we aim to explore the potential roles of FAM153B in GC. Microarray data were firstly obtained from public databases with the aim to evaluate the genetic expression of FAM153B between GC and normal tissues. The results were verified in immunohistochemistry (IHC). We also performed the co-expression network analysis and enrichment analysis to identify underlying mechanisms. A correlation analysis of FAM153B expression and immune infiltration was performed then. Furthermore, two GC cell lines were used to evaluate the effect of FAM153B on gastric cell proliferation by employing MTT and Edu assays. Our findings suggest that FAM153B is downregulated in tumoral tissue, and positively associated with unfavorable survival. The enrichment pathways of FAM153B were regulation of signaling receptor activity, DNA replication, cell cycle transition, chromosomal regulation, and so on. Besides, from the perspective of bioinformatics, the protein expression level of FAM153B is related to the degree of immune cell infiltration. In vitro, overexpression of FAM153B inhibit the proliferation of two cell lines. In summary, this study demonstrates that FAM153B might serve as an effective prognostic and therapeutic biomarker in GC.



    Gastric cancer (GC) is one of the most common digestive malignancies, and the incidence of GC gradually increased in recent years [1]. As the symptoms of early-stage GC may not be obvious, most patients with GC are detected in late and inoperable stages. Despite great progress in multimodal therapy, 5-year survival rates of patients with advanced GC are still less than 30% [2,3,4]. GC has emerged as a serious health problem and becomes a major burden worldwide, especially in East Asia [5,6]. Tumor biomarkers could serve as integral markers of pathogenic processes, disease activity, and therapeutic drug response. Advances in omic technologies have made more and more cancer-specific biomarkers been discovered, including DNA, RNA, exosome, etc. Rapid development in cancer biology has been tremendously promoted by the selection of appropriate biomarkers [7]. Distinct profiles in human cancers make it challenging in disease diagnosis, survival prediction, and treatment. Therefore, exploiting new molecular targets and prognostic biomarkers for GC become an urgent priority for disease management.

    Previously, we performed genome sequencing and bioinformatic analysis of gastric cancer tissues, thereby identifying several potential driver genes (data were not shown). Family with sequence similarity 153 member B (FAM153B) is a member of FAM153, and this family includes several coding proteins with unknown functions. FAM153A and FAM153C are the other two members of the FAM153 gene family. The FAM153B gene is located on chromosome 5 and its full-length cDNA is 1966 bp, which could encode protein (NP_0012525441). Its expression has been demonstrated in several monkeys and humans. Ferdouse et al. implemented a comprehensive genome-wide association study and demonstrated that FAM153B could be linked to lung disease [8]. However, the potential function and mechanism of FAM153B in cancers, especially gastric cancer, have not been reported. Thus, it is essential for comprehensive gene expression and functional analysis in GC. This will help elucidate diagnostic biomarker, prognostic indicator, and potential therapeutic target. Besides, this study also provides a rationale for further studies of gastric carcinogenesis.

    Combination of experimental and bioinformatics method contributed greatly to this subject. In this study, we firstly obtained the genetic data for GC from the public database, evaluated the differential expression and prognostic power of FAM153B through bioinformatics and experimental methods. To assess the potential function of FAM153B in GC, a series of bioinformation analysis, including immune analysis and pathway-enrichment analysis, were performed using online database and R software. Finally, two GC cell lines was utilized to investigate the potential biological function in vitro.

    Transcriptome data of FAM153B in multiple cancers and normal tissues were retrieved from the UCSC Xena browser (https://xenabrowser.net/datapages/). All the expression data were converted to TPM format, and log2 transformed. The log2 fold change was set to 1, while the P-value was set to 0.05.

    The receiving operating characteristics (ROC) curves were plotted to evaluate the diagnostic value. The area under the ROC curve were calculated by using the pROC package. AUC ≥ 0.70 was considered as better discrimination, and enabled us to make disease predictions.

    The procedure involving human data in this study was approved by the Ethics Committee of the First Hospital of Lanzhou University (LDYYLL2021-251). Tissue microarray analysis (TMA) was purchased from Shanghai Outdo Biotech, containing 65 GC and 65 adjacent stomach tissue. All patients were aged between 40 and 78 years. None of the patients received any treatment prior to surgery. All patients had at least 5 years of follow-up data (Table A1). Individuals with missing clinical events were excluded. All formalin-fixed paraffin-embedded (FFPE) tissues were cut into sections with the core size of 5 µm. Subsequently, these sections were stained with primary antibody against FAM153B (Solarbio, China). Primary antibody was used at a dilution of 1:150. FAM153B staining score was classified according to staining intensity and percentage of tumor cells. The staining intensity was classified as 0–3+ together with expression degree (no, weak, moderate, and strong). Percentage of positive tumor cells was characterized as 1 + (≤ 25%), 2 + (26–50%), 3 + (51–75%), and 4 + (>75%). The final score is defined by the product of the score from 0 to 12, and the median score of 6 was set as the threshold to distinguish between high- and low- expression. Finally, we analyzed the relationship between FAM153B expression and overall survival of these patients.

    The transcriptome data was firstly obtained from TCGA-STAD datasets. Subsequently, the modular analysis in LinkedOmics was performed to identify the co-expression genes of FAM153B in GC with FDR of 0.05. GSEA analysis was utilized to investigate the function of FAM153B. Besides, the top 50 positively/negatively correlated candidate genes were respectively used to construct the PPI network through GeneMINIA online analytical platform. Finally, the fundamental functions of these genes were explored with built-in functions of GeneMINIA platform.

    ssGSEA and GSVA were utilized to quantify twenty-four immune cell types. Subsequently, spearman correlation analysis was performed to explore the relationship between FAM153B expression and the composition of immune infiltrates.

    Human GC cell lines (MKN45and HGC27) were purchased from the American Type Culture Collection, and were cultured in DMEM and RP1640 media respectively, supplemented with 10% fetal bovine serum (FBS) and 1% antibiotics (streptomycin /penicillin). Cells were incubated at 37℃ in a 5% CO2 humidified environment.

    The cDNA fragment of human FAM153B (NM_001265615.2) was firstly synthesized and cloned into a lentiviral expression vector carrying a flag tag accordingly by Shanghai GeneChem Co., Ltd. The empty vector was used as a negative control. The cells were plated in a 6-well plate at 50–70 % confluency. Then, MKN45 and HGC27 cells were transduced with lentiviral particles carrying plasmids containing FAM153B or empty vector, respectively. The stably transfected cells were selected by using puromycin. The efficiency of FAM153B overexpression was confirmed by Western blot analysis.

    MTT and EdU were performed to evaluate the cell proliferation. The MTT Cell Proliferation and Cytotoxicity Assay Kit was purchased from Solarbio company. The cells were seeded into 96-well plates at a density of 5 × 103 cells/well. MTT dye (10 μl per well) was added into each well at different time points (0, 24, 48, and 72 h). The mixture was incubated for 4 h at 37 ℃. Next, the absorbance of wells at 490 nm was measured with a microplate reader (Thermo Scientific, United States). The EdU assay was performed by Meilun EdU Cell Proliferation Kit with Alexa Fluor 555 (Meilun, China) according to the manufacturer's instructions. The EdU cell line was photographed by using the Olympus IX51 microscope (Olympus, Japan).

    The total protein of GC cell lines was extracted by using RIPA buffer supplemented with phosphatase and protease inhibitors (Beyotime, China). Then, total proteins were collected and quantification was performed by using BCA kit (Solarbio, China). Total proteins were subjected to gel electrophoresis and sequentially transferred into a polyvinylidene fluoride (PVDF) membrane (Millipore, USA). After overnight incubation with primary antibodies (Invitrogen, USA), they were followed by incubating with a secondary antibody (Immunoway, USA) for 1 h. Primary antibody was used at a dilution of 1:1000. Membranes were developed with ECL substrate (Beyotime, China) and exposed using film.

    Graph Prism and R software were used for the statistical analysis. The differences between levels of FAM153B expression and clinical parameters were assessed by the chi-square test or Fisher-test. P-value less than 0.05 was considered as statistically significant.

    Based on TCGA and GTEx databases, we evaluated the level of FAM153B expression in pan-cancer tissues. FAM153B was found to be downregulated in 25 tumor tissues, but upregulated in CHOL, LAML, and THYM (Figure 1A). As shown in Figure 1B, FAM153B was downregulated approximately 1-fold in GC. To further evaluate the diagnostic value of FAM153B in GC, we performed the ROC analysis and calculated the AUC value. As shown in Figure 1C, FAM153B showed the promising diagnostic efficacy. To validate the expression levels of FAM153B in GC and normal tissues, we examined changes in protein levels of FAM153B from clinical samples using IHC. The results showed that the expression of FAM153B was lower in GC tissue than that of the normal tissue (Figure 2A, B).

    Figure 1.  Downregulation of FAM153B in GC tissues using public database. (A) The mRNA expression level of FAM153B in the TCGA-GTEx database was analyzed. (B) The gene expression level of FAM153B in the TCGA cohort dataset was analyzed. (C) Diagnostic value of FAM153B in GC. (* P < 0.05, **P < 0.01, ***P < 0.001).
    Figure 2.  Protein expression of FAM153B in GC tissues (Figure 2A) and adjacent non-tumor tissues (Figure 2B) were detected by IHC (n = 65).

    Kaplan-Meier curve was plotted to evaluate the association between overall survival and FAM153B expression. GC patients having lower expression of FAM153B showed a significantly reduced OS (P = 0.032, Figure 3). Based on their IHC scores of FAM153B in cancerous tissues, these patients were grouped into high (n = 15) and low (n = 50) expression. Statistical analysis revealed that the protein levels of FAM153B showed close association with GC patients' clinical parameter of overall survival (Table 1).

    Figure 3.  Kaplan-Meier analysis showed that the overall survival of GC patients with high or low FAM153B protein level.
    Table 1.  Relationship between FAM153B expression and clinical parameters of GC patients.
    Characteristics Low High P
    N = 65 N = 50 N = 15
    Age(years) ≤65 37 12 0.636
    > 65 13 3
    Gender MALE 36 11 0.919
    FEMALE 14 4
    Stage Ⅰ-Ⅱ 25 8 0.821
    Ⅲ-Ⅳ 25 7
    T T1-2 13 7 0.128
    T3-4 37 8
    N N0 14 3 0.536
    N1-3 36 12
    M M0 44 13 0.890
    M1 6 2
    Differentiation well-moderately differentiated 20 8 0.506
    poorly differentiated 26 7
    unknown 4 0
    Tumor size ≥ 5 cm 14 4 0.758
    < 5 cm 33 14
    Tumor location Proximal + middle 15 5 0.746
    Distal 32 13
    Treatment Proximal + total gastrectomy 16 3 0.229
    distal gastrectomy 31 15
    Helicobacter pylori Negative 16 7 0.262
    Positive 24 5
    Unknown 10 3
    HER2 status Negative 42 15 0.321
    Positive 6 0
    Unknown 2 0
    Survival Alive 14 9 0.023
    Dead 36 6

     | Show Table
    DownLoad: CSV

    In order to elucidate the biological significance of FAM153B, the change in co-expression of genes was obtained, and further involved in pathway enrichment analysis. We identified a total of 7084 positively correlated genes and 638 negatively correlated genes with statistically significant difference (FDR < 0.05). As shown in Figure 4, we presented a heatmap of the top 50 most relevant positive and negative genes, respectively. GSEA analysis indicated that these genes were involved in DNA replication-related pathway. The PPI network constructed around positive/negative genes reveals similarity between these genes. The PPI network of positive genes showed an enrichment in signaling activity-related genes, while the PPI network of negative genes showed an enrichment in DNA replication-related genes.

    Figure 4.  FAM153B co-expression genes and its relevant regulatory pathways in GC. (A, B) Heat maps show the top 50 positive and top 50 negative correlated genes of FAM153B in GC. (C, D) The biological functions of positive and negative correlated genes were evaluated using PPI network based on GeneMINIA.

    The correlation between FAM153B expression and immune cell infiltration levels was determined by Spearman correlation analysis. As shown in Figure 5, the expression of FAM153B was positively correlated with immune cells (NK cells, TFH, B cells, Tcm) (P < 0.05), and negatively correlated with (Neutrophils, NK CD56dim cells, Th17 cells, Th2 cells) (P < 0.05). Analysis of FAM153B expression and corresponding immune cells indicated that FAM153B was significantly negatively associated with Th2 expression (P < 0.05).

    Figure 5.  The expression level of FAM153B was correlated with immune cells infiltration in GC. (A) Correlation between the proportions of different immune cells and FAM153B expression levels. (B) The histograms of immune cells infiltration level related to the expression level of FAM153B. (C–J) Spearson's analysis showing the difference of immune cells infiltration level based on expression of the candidate protein.

    We firstly examined endogenous expression of FAM153B in six GC cell lines mentioned in method part. Six GC cell lines expressed FAM153B in various degrees (Figure A1). The protein expression levels of FAM153B were relatively lower in MKN45 and HGC27 cells. Two cell lines were selected for further lentivirus transfection. The FAM153B overexpression group and control group were named Control and OE-FAM153B respectively. Western blotting indicated that protein expression of FAM153B was notably increased in OE-FAM153B GC cells compared with that in the control vector (Figure 6A, B). We applied MTT and EdU separately to investigate the effect of FAM153B on cell proliferation. Different degrees of cell proliferation inhibition were observed in MKN45 cell lines at 48 and 72 h, HGC27 cell lines at 24, 48 and 72 h (Figure 6C, D). Next, the results of EdU staining showed that the proliferation of MKN45 and HGC27 cells was moderately reduced in OE-FAM153B group as compared with vector group (Figure 6E, F). In summary, these results suggested that the overexpression of FAM153B could attenuate GC cell proliferation.

    Figure 6.  FAM153B inhibits gastric cancer growth in vitro. (A, B) Western blotting showing successful overexpression of FAM153B in gastric cancer cell lines (MKN45, HGC27). (C, D) MTT assays were performed to evaluate cell viability, indicating that two cells overexpressing FAM153B showed the repression of proliferation capacity more obviously. (E, F) Proliferation of MKN45 and HGC27 cells were evaluated by EdU labeling assay and showed that overexpressing FAM153B led to proliferation inhibitions of proliferation capacity.

    To our knowledge, this is the first study to explore the potential effects of FAM153B in GC. This study revealed that FAM153B was downregulated in GC tissues, indicating that FAM153B has a potential impact on GC. We confirmed the low expression of FAM153B in GC via public database analysis. This observation was further validated by an IHC analysis in our independent cohort. The clinical importance of FAM153B in GC is highlighted by the correlation of lower FAM153B with adverse prognosis. To confirm the clinical relevance of FAM153B, we analyzed the relationship between FAM153B expression and clinical parameters of GC patients. The fact that a statistically significant difference between both FAM153B-low and -high group further supports its potential clinical relevance. Although the groups differed in several clinical variables, including age, gender, stage, Helicobacter pylori, the treatments, etc., these factors did not exhibit statistically significant differences. These results hint at more favorable aspects of prognosis prediction.

    The pathological function of FAM153B is rarely explored in different diseases. Yet, until now only Ferdouse et al. have reported the preliminary mechanism in COPD [8]. The biological role of FAM153B in GC have not been studied. Therefore, FAM153B was selected and underwent evaluation of identifying its possible mechanism in GC. We sought its clinical significance and speculated that FAM153B has a function as the tumor suppressor gene for GC. Gene co-expression network analysis is commonly used to investigate the genes of unknown function and understand its interactions. Recent advancement in transcriptome contributed to the construction of these networks, and further infer the functions of genes. To further investigate the underlying function of FAM153B in GC, we performed the co-expression network analysis and enrichment analysis. Our results revealed enrichment of these co-expressed genes of FAM153B in primary pathways related to DNA replication. DNA replication is the fundamental physiological process that ensure accurate transfer of genetic information [9]. Dysregulation of DNA replication could cause genome instability, which was considered as a hallmark of cancer [10]. Previous studies indicated that DNA replication perturbations could have influences on the replicative stress, and been induced at the early steps of cancer development [11]. This stress could not only trigger genome instability, but also cause chromosomal instability that further generate intratumoral heterogeneity in response to external pressures [11]. Accumulating evidences suggest that several cancer cells, including gastric cancer, rely on DNA replication stress related pathways for promoting its proliferation [12,13,14,15]. After the transfection of FAM153B to MKN45 and HGC27 cell lines, we found cell proliferation suppression in the two cell lines. Edu proliferation assay could directly and accurately detect the synthesis of DNA. Therefore, we demonstrated that FAM153B was associated with DNA replication [16]. However, the specific molecular mechanism deserves further exploration.

    As we all known, endogenous processes such as DNA replication could affect genome instability, thus interfering with the immunogenicity [17]. Immune cell infiltration was closely related to the immunogenicity. Growing evidences have demonstrated that the immune cell infiltration affects cancer cell viability and is associated with disease prognosis [18,19]. In the solid tumor microenvironment, the presence of dynamic tumor-immune cell interaction causes the release of cytokines and growth factors [20]. Such crosstalk could create the necessary conditions for tumor growth and metastasis [20]. Previous study suggested that Th2-like cells could orchestrate malignant phenotypes of melanoma and colorectal cancer, and might contribute to the pro-tumorigenic environment [21]. A recent study by Lan et al. found that high levels of Th2 cells are correlated with aggressive characteristics in breast cancer, especially ER-positive breast cancer [22]. The results from Schreiber S. et al elaborate unique correlations between Th2 cells and the tumor microenvironment, thereby revealing novel avenues to immunotherapies [23]. Besides, Th2 cell-related pathway has been proven to contribute to H. pylori-associated gastric tumourigenesis or intestinal metaplasia [24]. Therefore, Th2 cell might participate in the carcinogenesis process. In this study, we uncovered a negative correlation between Th2 cell and the expression level of FAM153B. Besides, our results indicate that overexpression of FAM153B could inhibit the proliferation of gastric cancer cells. In this process, Th2 cell, as primary immune cell, might be regulated by FAM153B. However, further studies are needed to confirm this inference.

    Although FAM153B might act as a tumor suppressor gene in GC, there are certain limitations in this study. First, we only performed in vitro experiments to explore the potential function of FAM153B. In vivo studies were not conducted accordingly. Second, a limited number of clinical specimens were available in this study. Strict inclusion and exclusion criteria offered advantages to the collection of samples. However, future studies need to collect more samples for the exploration of correlation between FAM153B expression and relevant clinical variables, including prognostic features. Third, specific mechanisms of DNA replication and immune cell infiltration on FAM153B remain to be fully elucidated. As we now know, the occurrence and development of GC is a complex biological process involving multiple factors, and there exists various regulatory mechanisms. Therefore, further studies are warranted to provide the detailed analysis of FAM153B, to explore the underlying regulatory mechanism in tumorigenesis of GC.

    In summary, this study demonstrated that FAM153B might serve as an effective prognostic biomarker and function as a tumor suppressor gene in GC.

    This study was supported by the Natural Science Foundation of Gansu province (20JR10RA685, 21JR11RA099, 21JR1RA117) and the Foundation of The First Hospital of Lanzhou University (ldyyyn2018-38, ldyyyn2020-95).

    The authors report no conflicts of interest in this work.

    Figure A1.  The expression of FAM153B in gastric cancer cell lines.
    Table A1.  Clinical information of patients with gastric cancer.
    Id gender age surgery date fustat futime treatment location stage T N M differentiation HER2 Hp
    67186-3 male 66 2015.2.2 death 21 total gastrectomy antrum T4N3M0 4 3 0 poorly adenocarcinoma +++ unknown
    64549-3 male 57 2014.8.1 death 37 proximal gastrectomy fundus T3N1M0 3 1 0 moderately adenocarcinoma - (+)
    64394-3 male 67 2014.8.6 death 39 proximal gastrectomy fundus T3N2M0 3 2 0 poorly adenocarcinoma - (-)
    65922-2 male 71 2014.12.1 death 11 proximal gastrectomy body T4N2M0 4 2 0 moderately adenocarcinoma +++ (-)
    63061-3 male 72 2014.5.4 alive 60 proximal gastrectomy fundus T3N3M0 3 3 0 moderately adenocarcinoma +++ unknown
    62982-3 male 57 2014.4.21 death 26 proximal gastrectomy body T4N2M0 4 2 0 poorly adenocarcinoma unknown
    62019-3 male 70 2014.2.27 alive 60 proximal gastrectomy fundus T3N0M0 3 0 0 poorly adenocarcinoma - unknown
    69540-4 male 62 2015.7.2 death 32 proximal gastrectomy fundus T3N2M0 3 2 0 poorly adenocarcinoma - (+)
    69953-1 male 70 2015.7.27 death 18 proximal gastrectomy fundus T4N2M0 4 2 0 poorly adenocarcinoma - (+)
    70335-4 male 55 2015.7.10 alive 60 proximal gastrectomy fundus T2N1M0 2 1 0 moderately adenocarcinoma - (-)
    68012-3 male 63 2015.3.24 death 56 proximal gastrectomy fundus T4N0M0 4 0 0 poorly adenocarcinoma - unknown
    67381-3 female 51 2015.2.9 death 11 total gastrectomy body T4N3M1 4 3 1 poorly adenocarcinoma - (-)
    67527-4 female 54 2015.2.25 alive 60 total gastrectomy fundus T3N3M0 3 3 0 poorly adenocarcinoma - (+)
    62249-3 male 64 2014.3.5 death 15 total gastrectomy body T2N0M0 2 0 0 poorly adenocarcinoma - (-)
    66530-3 male 54 2015.1.27 death 10 total gastrectomy antrum T3N1M0 3 1 0 moderately adenocarcinoma - (-)
    69117-3 female 59 2015.6.4 death 2 total gastrectomy body T3N3M0 3 3 0 moderately adenocarcinoma - (+)
    64747-3 male 60 2014.8.13 death 47 total gastrectomy body T3N2M0 3 2 0 poorly adenocarcinoma - (+)
    69928-1 female 53 2015.7.24 alive 60 total gastrectomy body T4N3M0 4 3 0 moderately adenocarcinoma - unknown
    66999-3 male 50 2015.1.24 death 33 total gastrectomy fundus T3N3M0 3 3 0 poorly adenocarcinoma - unknown
    63356-3 male 64 2014.5.14 death 10 distal gastrectomy antrum T4N3M1 4 3 1 mucinous adenocarcinoma - (-)
    67039-2 male 58 2015.1.26 alive 60 distal gastrectomy antrum T3N1M0 3 1 0 moderately adenocarcinoma - (+)
    67080-3 male 62 2015.1.27 death 10 distal gastrectomy antrum T4N3M1 4 3 1 moderately adenocarcinoma - (-)
    62546-3 female 62 2014.3.21 alive 60 distal gastrectomy antrum T2N2M0 2 2 0 moderately adenocarcinoma - (+)
    61283-4 female 65 2014.1.8 death 30 distal gastrectomy antrum T4aN0M0 4a 0 0 moderately adenocarcinoma +++ (-)
    66364-2 female 59 2014.12.15 death 38 distal gastrectomy antrum T2N3M0 2 3 0 moderately adenocarcinoma - (+)
    63128-3 male 47 2014.4.29 alive 60 distal gastrectomy antrum T3N0M0 3 0 0 moderately adenocarcinoma - unknown
    61155-3 male 42 2013.12.31 death 36 distal gastrectomy antrum T3N3M1 3 3 1 poorly adenocarcinoma - (+)
    62927-3 male 54 2014.4.17 death 58 distal gastrectomy antrum T2N0M0 2 0 0 moderately adenocarcinoma - (+)
    66174-3 male 56 2014.12.2 alive 60 distal gastrectomy antrum T1N1M0 1 1 0 highly
    adenocarcinoma
    - (+)
    64144-3 male 67 2014.7.10 death 25 distal gastrectomy antrum T3N3M1 3 3 1 poorly adenocarcinoma - (+)
    65581-3 male 68 2014.10.15 death 31 distal gastrectomy antrum T3N3M0 3 3 0 poorly adenocarcinoma - (+)
    65731-3 male 69 2014.10.28 death 23 distal gastrectomy antrum T4N3M0 4 3 0 moderately adenocarcinoma +++ (+)
    63432-3 male 63 2014.5.21 death 40 distal gastrectomy antrum T3N0M0 3 0 0 mucinous adenocarcinoma +++ (+)
    61668-3 male 53 2014.1.27 death 16 distal gastrectomy antrum T3N0M0 3 0 0 poorly adenocarcinoma - unknown
    61817-3 male 48 2014.2.13 alive 60 distal gastrectomy antrum T2N1M0 2 1 0 poorly adenocarcinoma - (-)
    62082-3 male 54 2014.2.27 alive 60 distal gastrectomy antrum T2N1M0 2 1 0 poorly adenocarcinoma - (+)
    62760-3 male 57 2014.4.8 death 42 distal gastrectomy antrum T3N3M0 3 3 0 poorly adenocarcinoma - (+)
    63163-3 female 46 2014.5.4 alive 60 distal gastrectomy antrum T2N0M0 2 0 0 poorly adenocarcinoma - unknown
    63321-2 female 63 2014.5.21 death 28 distal gastrectomy antrum T4N1M0 4 1 0 moderately adenocarcinoma - (-)
    63489-3 male 61 2014.5.26 alive 60 distal gastrectomy antrum T3N2M0 3 2 0 moderately adenocarcinoma - unknown
    64067-4 male 40 2014.7.4 death 27 distal gastrectomy antrum T3N2M0 3 2 0 poorly adenocarcinoma - unknown
    64528-2 female 56 2014.7.31 death 20 distal gastrectomy antrum T4N3M1 4 3 1 poorly adenocarcinoma - (+)
    65313-3 male 73 2014.9.19 alive 60 distal gastrectomy antrum T3N0M0 3 0 0 poorly adenocarcinoma - (+)
    65405-3 male 51 2014.9.29 death 39 distal gastrectomy antrum T3N3M0 3 3 0 poorly adenocarcinoma - (+)
    66135-3 male 43 2014.11.28 death 35 distal gastrectomy antrum T3N0M0 3 0 0 poorly adenocarcinoma - (-)
    66541-3 female 41 2014.12.26 alive 60 distal gastrectomy antrum T1N1M0 1 1 0 poorly adenocarcinoma - (+)
    66972-3 female 67 2015.1.23 death 41 distal gastrectomy body T3N1M0 3 1 0 mucinous adenocarcinoma - (-)
    63528-3 male 58 2014.5.28 alive 60 distal gastrectomy antrum T2N2M0 2 2 0 poorly adenocarcinoma - (+)
    66457-3 male 40 2015.1.26 alive 60 distal gastrectomy antrum T1N1M0 1 1 0 moderately adenocarcinoma - (+)
    69284-3 female 60 2015.6.15 alive 60 distal gastrectomy antrum T1N0M0 1 0 0 moderately adenocarcinoma - (-)
    70634-3 female 68 2015.9.10 death 30 distal gastrectomy antrum T3N0M0 3 0 0 moderately adenocarcinoma - (+)
    70708-3 female 68 2015.9.16 death 50 distal gastrectomy antrum T1N2M0 1 2 0 highlyadenocarcinoma - (-)
    68762-3 male 60 2015.5.13 death 58 distal gastrectomy antrum T1NOM0 1 0 0 highlyadenocarcinoma - (-)
    70411-3 female 69 2015.8.24 death 31 distal gastrectomy antrum T3N2M0 3 2 0 moderately adenocarcinoma - (-)
    68634-2 male 45 2015.5.5 alive 60 distal gastrectomy antrum T1N0M0 1 0 0 moderately adenocarcinoma - (-)
    68054-3 male 58 2015.3.26 death 9 distal gastrectomy antrum T2N3M1 2 3 1 poorly adenocarcinoma - (+)
    68131-3 female 76 2015.4.1 death 31 distal gastrectomy antrum T1bN2M0 1 2 0 poorly adenocarcinoma - (-)
    68354-3 female 60 2015.4.15 death 13 distal gastrectomy antrum T4N3M1 4 3 1 poorly adenocarcinoma - (-)
    69285-3 male 57 2015.6.15 death 38 distal gastrectomy antrum T3N1M0 3 1 0 poorly adenocarcinoma - (+)
    70841-2 male 62 2015.9.24 alive 60 distal gastrectomy antrum T3N0M0 3 0 0 moderately adenocarcinoma - (-)
    70982-3 male 49 2015.10.12 alive 60 distal gastrectomy body T4N2M0 4 2 0 moderately adenocarcinoma - (+)
    71194-3 male 60 2015.10.29 alive 60 distal gastrectomy antrum T2N2M0 2 2 0 poorly adenocarcinoma - (-)
    69050-4 male 65 2015.5.24 alive 60 distal gastrectomy antrum T2N0M0 2 0 0 poorly adenocarcinoma - (+)
    71357-3 male 78 2015.11.11 death 13 distal gastrectomy antrum T4N3M0 4 3 0 moderately adenocarcinoma (-)
    68518-3 male 48 2015.4.23 death 45 distal gastrectomy antrum T3N3M0 3 3 0 mucinous adenocarcinoma - unknown

     | Show Table
    DownLoad: CSV


    [1] H. Sung, J. Ferlay, R. L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal, et al., Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries, CA: A Cancer J. Clin., 71 (2021), 209–249. https://doi.org/10.3322/caac.21660 doi: 10.3322/caac.21660
    [2] R. E. Sexton, M. N. Al Hallak, M. Diab, A. S. Azmi, Gastric cancer: a comprehensive review of current and future treatment strategies, Cancer Metastasis Rev., 39 (2020), 1179–1203. https://doi.org/10.1007/s10555-020-09925-3 doi: 10.1007/s10555-020-09925-3
    [3] M. Banks, D. Graham, M. Jansen, T. Gotoda, S. Coda, M. di Pietro, et al., British Society of Gastroenterology guidelines on the diagnosis and management of patients at risk of gastric adenocarcinoma, BMJ, 68 (2019), 1545–1575. https://doi.org/10.1136/gutjnl-2018-318126 doi: 10.1136/gutjnl-2018-318126
    [4] A. Zayac, K. Almhanna, Esophageal, gastric cancer and immunotherapy: small steps in the right direction, Transl. Gastroenterol. Hepatol., 5 (2020), 9. https://doi.org/10.21037/tgh.2019.09.05 doi: 10.21037/tgh.2019.09.05
    [5] A. P. Thrift, H. B. El-Serag, Burden of gastric cancer, Clin. Gastroenterol. Hepatol., 18 (2020), 534–542. https://doi.org/10.1016/j.cgh.2019.07.045 doi: 10.1016/j.cgh.2019.07.045
    [6] M. Cao, H. Li, D. Sun, W. Chen, Cancer burden of major cancers in China: a need for sustainable actions, Cancer Commun., 40 (2020), 205–210. https://doi.org/10.1002/cac2.12025 doi: 10.1002/cac2.12025
    [7] T. Matsuoka, M. Yashiro, Biomarkers of gastric cancer: current topics and future perspective, World J. Gastroenterol., 24 (2018), 2818–2832. https://doi.org/10.3748/wjg.v24.i26.2818 doi: 10.3748/wjg.v24.i26.2818
    [8] B. Ferdouse, I. Ruczinski, S. Li, E. Silverman, M. Cho, D. Lynch, et al., Identifying a deletion affecting total lung capacity among subjects in the COPDGene study cohort, Genet Epidemiol, 1 (2016), 81–88. https://doi.org/10.1002/gepi.21943 doi: 10.1002/gepi.21943
    [9] M. Macheret, T. D. Halazonetis, DNA replication stress as a hallmark of cancer, Annu. Rev. Pathol.: Pathol. Mech. Dis., 10 (2015), 425–448. https://doi.org/10.1146/annurev-pathol-012414-040424 doi: 10.1146/annurev-pathol-012414-040424
    [10] H. Zhu, U. Swami, R. Preet, J. Zhang, Harnessing DNA replication stress for novel cancer therapy, Genes, 11 (2020), 990. https://doi.org/10.3390/genes11090990 doi: 10.3390/genes11090990
    [11] H. Kitao, M. Iimori, Y. Kataoka, T. Wakasa, E. Tokunaga, H. Saeki, et al., DNA replication stress and cancer chemotherapy, Cancer Sci., 109 (2018), 264–271. https://doi.org/10.1111/cas.13455 doi: 10.1111/cas.13455
    [12] J. Shi, X. Zhang, J. Li, W. Huang, Y. Wang, Y. Wang, et al., MTA2 sensitizes gastric cancer cells to PARP inhibition by induction of DNA replication stress, Transl. Oncol., 14 (2021), 101167. https://doi.org/10.1016/j.tranon.2021.101167 doi: 10.1016/j.tranon.2021.101167
    [13] M. Bauer, Z. Nascakova, A. I. Mihai, P. F. Cheng, M. P. Levesque, S. Lampart, et al., The ALPK1/TIFA/NF-κB axis links a bacterial carcinogen to R-loop-induced replication stress, Nat. Commun., 11 (2020), 5117. https://doi.org/10.1038/s41467-020-18857-z doi: 10.1038/s41467-020-18857-z
    [14] N. S. Sethi, O. Kikuchi, G. N. Duronio, M. D. Stachler, J. M. McFarland, R. Ferrer-Luna, et al., Early TP53 alterations engage environmental exposures to promote gastric premalignancy in an integrative mouse model, Nat. Genet., 52 (2020), 219–230. https://doi.org/10.1038/s41588-019-0574-9 doi: 10.1038/s41588-019-0574-9
    [15] J. Rozacky, A. A. Nemec, J. B. Sweasy, D. Kidane, Gastric cancer associated variant of DNA polymerase beta (Leu22Pro) promotes DNA replication associated double strand breaks, Oncotarget, 6 (2015), 24474–24487. https://doi.org/10.18632/oncotarget.4426 doi: 10.18632/oncotarget.4426
    [16] H. Hua, S. E. Kearsey, Monitoring DNA replication in fission yeast by incorporation of 5-ethynyl-2'-deoxyuridine, Nucleic Acids Res., 39 (2011), e60. https://doi.org/10.1093/nar/gkr063 doi: 10.1093/nar/gkr063
    [17] E. R. Mardis, Neoantigens and genome instability: impact on immunogenomic phenotypes and immunotherapy response, Genome Med., 11 (2019), 71. https://doi.org/10.1186/s13073-019-0684-0 doi: 10.1186/s13073-019-0684-0
    [18] D. Bruni, H. K. Angell, J. Galon, The immune contexture and immunoscore in cancer prognosis and therapeutic efficacy, Nat. Rev. Cancer, 20 (2020), 662–680. https://doi.org/10.1038/s41568-020-0285-7 doi: 10.1038/s41568-020-0285-7
    [19] W. H. Fridman, L. Zitvogel, C. Sautès-Fridman, G. Kroemer, The immune contexture in cancer prognosis and treatment, Nat. Rev. Clin. Oncol., 14 (2017), 717–734. https://doi.org/10.1038/nrclinonc.2017.101 doi: 10.1038/nrclinonc.2017.101
    [20] S. Y. Gun, S. W. L. Lee, J. L. Sieow, S. C. Wong, Targeting immune cells for cancer therapy, Redox Biol., 25 (2019), 101174. https://doi.org/10.1016/j.redox.2019.101174 doi: 10.1016/j.redox.2019.101174
    [21] L. Halim, M. Romano, R. McGregor, I. Correa, P. Pavlidis, N. Grageda, et al., An atlas of human regulatory T helper-like cells reveals features of Th2-like tregs that support a tumorigenic environment, Cell Rep., 20 (2017), 757–770. https://doi.org/10.1016/j.celrep.2017.06.079 doi: 10.1016/j.celrep.2017.06.079
    [22] L. Le, Y. Tokumaru, M. Oshi, M. Asaoka, L. Yan, I. Endo, et al., Th2 cell infiltrations predict neoadjuvant chemotherapy response of estrogen receptor-positive breast cancer, Gland Surg., 10 (2021), 154–165. https://doi.org/10.21037/gs-20-571 doi: 10.21037/gs-20-571
    [23] S. Schreiber, C. M. Hammers, A. J. Kaasch, B. Schraven, A. Dudeck, S. Kahlfuss, Metabolic interdependency of Th2 cell-mediated type 2 immunity and the tumor microenvironment, Front. Immunol., 12 (2021), 632581. https://doi.org/10.3389/fimmu.2021.632581 doi: 10.3389/fimmu.2021.632581
    [24] B. Marotti, A. Rocco, P. De. Colibus, D. Compare, G. de. Nucci, S. Staibano, et al., Interleukin-13 mucosal production in Helicobacter pylori-related gastric diseases, Dig. Liver Dis., 40 (2008), 240–247. https://doi.org/10.1016/j.dld.2007.11.02 doi: 10.1016/j.dld.2007.11.02
  • Reader Comments
  • © 2022 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(2422) PDF downloads(81) Cited by(0)

Figures and Tables

Figures(7)  /  Tables(2)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog