Introduction
Esophageal squamous cell carcinoma (ESCC) is a very common human cancer, and ranked 6th most common cancer worldwide. In Taiwan, it is the 5th leading cause of death among men, particularly prevalent in South-East and Central Asia. Esophageal cancer is histologically classified as either squamous cell carcinoma or adenocarcinoma [1]. Surgical resection is the main-stream treatment for early-stage esophageal carcinoma. For locally advanced esophageal carcinoma. The approach of multidisciplinary therapy, like radiotherapy, chemotherapy, and surgery, has been developed to prolong the patient survival. Despite this, the prognosis remains poor [2, 3]. Previous studies reported that Neoadjuvant chemoradiotherapy (nCRT) followed by surgery is a common multidisciplinary treatment for resectable esophageal carcinoma [4–11]. But the prognosis remains disappointing, with >50% of patients showing poor response to nCRT [12–15]. No simple and reliable criterion is available currently to determine the success of such therapy. According to the clinicopathologic and gene-expression profiles, some studies reported that tumor size and molecular makers, such as ERCC1, GNAS T93C, ABCB1 C3435T, might be associated with the response of chemotherapy or radiotherapy [8, 16–20]. These studies are mainly based on the post-treatment specimen as treatment-naïve specimens before nCRT are not available. Therefore, it is difficult to apply in clinical practice. Here, we aim to identify biomarkers from treatment-naïve specimens that allow early prediction the response to nCRT. Results would be useful to develop alternative personalized therapy or targeted therapy based on biomarkers.
As broad tumor profiling becomes a common component of cancer care, next-generation sequencing (NGS) is increasingly used in many areas of cancer research and clinical settings. Furthermore, endoscopic biopsies are suitable for targeted NGS, which provides quality sequencing data and accurate information on mutations [21–23]. NGS is a tool that is also widely available to gastroenterologists [21–23]. In this study, we aimed to identify potential genes for predicting response to therapy based on NGS biopsy samples from ESCC patients. Based on function analysis results, we chose AGR2 to perform further investigation. Results showed that silencing AGR2 enhances sensitivity to the cytotoxic effects of cisplatin and 5-fluorouracil (5-FU). We concluded that AGR2 is a potential gene marker for predicting response to ESCC therapy.
Materials and methods
Patient selection
From January 1, 2016, to December 31, 2018, we retrospectively enrolled 32 ESCC patients who had undergone nCRT at the Taichung Veterans General Hospital. These patients each had one endoscopic specimen of pre-treatment biopsy (treatment-naïve tissue) and another specimen of post-treatment biopsy. Samples of surgically resected tumors after nCRT were obtained from the Biobank of Taichung Veterans General Hospital. We collected their clinical information such as age, sex, surgery type, complete or incomplete resection, histologic subtype, tumor stage, clinical image data, and therapeutic response. Both the data collection procedure and the gene expression analysis of tumor tissues were approved by the Institutional Review Board of Taichung Veterans General Hospital (IRB TCVGH No: CE17279A). All patients did not contain minors and other vulnerable groups provided written informed consent to participate in this study before enrollment.
RNA sequencing and gene expression analysis
RNA libraries were generated using the TruSeq Stranded mRNA Library Prep Kit (Illumina, SanDiego, CA, USA) with 1μg of total RNA from all samples following the manufacturer’s instructions. The prepared library was sequenced with paired-end runs using the Illumina HiSeq 2500 sequencer. RNA reads were mapped onto the human reference genome GRCh37 using the HISAT2aligner tool [24]. Read counts were calculated using feature Counts [25] and gene expression profiles were identified using DESeq2 [26]. The DAVID functional tool (the Database for Annotation, Visualization, and Integrated Discovery, https://david.ncifcrf.gov/) was used for functional annotation of differentially expressed genes. The Metascape (http://metascape.org/gp/index.html#/main/step1) online tools were used to analyze Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment [27].
Cell line and culture conditions
We used two esophageal cancer cell lines (CE48T/VGH, and CE146T/VGH). Each was cultured in Dulbecco’s modified Eagle’s medium (Gibco, Grand Island, NY, USA) supplemented with 2mM L-glutamine (Gibco, Grand Island, NY, USA), 10% fetal bovine serum (Gibco, Grand Island, NY, USA), 100 U/mL penicillin-streptomycin (Gibco, Grand Is-land, NY, USA), and 10mM non-essential amino acids (Gibco, Grand Island, NY, USA). TE-2 cells were cultured in the same medium supplemented with 1mM sodium pyruvate (Gib-co, Grand Island, NY, USA). All cells were cultured in a 5% CO2 atmosphere at 37°C.
RNA interference (small interfering RNA) analysis
The RNA interference (RNAi) technology has revolutionized biological discovery, target discovery, and validation processes. A SmartPool of 4 siRNA sequences derived from the coding sequence of AGR2 and individual duplex and control siRNA were designed and purchased from Dharmacon (Lafayette, CO, USA). The following siRNAs were used: AGR2 siRNA no.1: 5′—GCUGAAGACUGAAUUGUAA-3′, no.2: 5’-GCAACAAACCCUUGAUGAU-3′, no.3: AGUCAAACCUGGAGCCAAA-3′, and no.4 5′-UGAAGAAAGCUCUCAAGUU-3′. The control siRNA was non-targeting pool sequences that included the following: no. 1: 5′-UGGUUUACAUGUCGACUAA-3′, no. 2: 5′-UGGUUUACAUGUUGUGUGA-3′, no. 3: 5′-UGGUUUACAUGUUUUCUGA-3′, and no. 4: 5′-UGGUUUACAUGUUUUCCUA-3′. Each freeze-dried siRNA was dissolved in RNase-free water.
Using siRNA, we knocked down AGR2 gene expression in esophageal cancer cells. The TransIT-X2 Dynamic Delivery System reagent (Mirus Bio, Madison, WI, USA) procedure was used to forward-transfect siRNA into the esophageal cancer cells. The esophageal cancer cell cells were put in 6-well culture plates at a density of 4.0–6.0 ×105 cells/well, and cultured in 2 mL growth medium for 24 hr. Cells were transfected with siRNA to a final concentration of 25 nM as diluted with the TransIT-X2 transfection reagent. Subsequently, cells were incubated with 5% CO2 at 37°C for 72 hr. Finally, cells were harvested and assayed for the knockdown of target gene expression.
Reverse transcription and quantitative polymerase chain reaction
The total RNA was extracted using the AllPrep DNA/RNA Mini Kit, following the manufacturer’s protocol (cat. 80204). Reverse transcription was done using the SuperScriptTM IV Reverse Transcriptase protocol (Invitrogen, Vilnius, Lithuania). Quantitative reverse-transcription polymerase chain reaction was done using the FastStart TaqMan Probes system (Cat.4913947001, Roche Diagnostics, Indianapolis, IN, USA) with AGR2 specific primers. The analysis was performed on a StepOne Plus Real‐Time PCR System (Applied Biosystems, Foster, CA). Glyceraldehyde 3-phosphate dehydrogenase was used as endogenous control to quantify determine the relative expression levels of target genes using the2-ΔΔct method.
Reagents
Cisplatin (P4394) and 5-fluorouracil (F6627) were purchased from Sigma-Aldrich, ST Louis, MO. Cisplatin was dissolved in double-distilled water. 5-FU was dissolved in dimethyl sulfoxide (DMSO) (Sigma-Aldrich, ST Louis, MO). Same solvent was used in the control experiment.
MTT assay
To determine the cytotoxicity of the combined effect of AGR2 knockdown and chemotherapeutic agents, cells were first put in 24-well culture plates for 24 hr. Then, cells were transfected with siRNA. After 24 hr, we treated the cells with cisplatin (2.0–6.0μM) and 5-FU (3.0–20.0μM) for 72hr. Cell viability was evaluated using the MTT assay. The medium was removed and cells were washed twice with phosphate-buffer saline (PBS). Then, 500 μL MTT solutions (1mg/mL) (Biomatik, Ontario, Canada) were added, and preparations incubated at 37°C for 30 min. The MTT solution was removed and replaced with 200 μL DMSO. Subsequently, cells were incubated for 5 min. We transferred 100 μL DMSO of dissolved cells into 96-well enzyme-linked immunosorbent assay (ELISA) plates to measure absorbance at 570/670 nm using an ELISA reader. Each experimental data point represents the average value of three replicates.
Annexin V/propidium iodide apoptosis assay
The Annexin V-FITC Apoptosis Detection Kit was purchased from BioVision. Annexin V and propidium iodide (PI) double staining was performed following the manufacturer’s instructions. After staining, cells were analyzed with the flow cytometry.
Western blotting
The western blot was used to determine levels of AGR2 and associated proteins. Cells were first washed with PBS and lysed in RIPA lysis buffer (APOLO, Hsinchu, Taiwan) containing 50mM Tris, 0.15M NaCl, 1% NP-40, 0.5% sodium deoxycholate, and 0.1% SDS supplemented with a protease inhibitor cocktail (MCE, Monmouth Junction, NJ, USA). Protein concentrations were detected using a protein assay kit (Bio-Rad, Hercules, CA, USA). Equal amounts of proteins (30 μg) were subjected to sodium dodecyl sulfate 8% -12% polyacrylamide gel electrophoresis. Fractionated proteins were transferred to Hybond-P PVDF membranes (Millipore, Darmstadt, DE). Membranes were blocked with PBS containing 5% nonfat milk and 0.2% Tween 20. For the detection of human anti-AGR2 (Invitrogen, Vilnius, Lithuania) and anti-β-actin (Sigma-Aldrich, St Louis, MO), the membranes were incubated overnight at 4°C, followed by the addition of anti-mouse IgG or anti-rabbit IgG antibody linked to Horseradish peroxidase (Jackson, West Grave, PA, USA). Blots were finally developed using an enhanced chemiluminescence reagent (Millipore, Darmstadt, DE).
Statistical analyses
Statistical analyses were performed using the paired two-way analysis of variance with Tukey’s test. All results reflect the mean ± standard error of the mean data obtained from at least three independent experiments. Statistical was set defined as p <0.05.
Results
Clinical characteristic
We collected 32 patients with ESCC. Their clinicopathological characteristics are summarized in Table 1. Standard protocols for patients with operable esophageal cancer are bridfed as follows. Chemotherapy is given concurrently with cisplatin 20 mg/ml iv for 1 hour and fluorouracil 800 mg/ml iv for 24 hours on a daily basis from day 1 to 4 (cycle1), and from day 29 to 32 (cycle 2) with radiotherapy. Radiotherapy is performed 5 days per week, with a daily dose of 180 Gy over a total course of 5 to 6 weeks. Surgery was performed 4 to 6 weeks after completing nCRT. The procedure included thoracoscopic esophagectomy, at least 2-eld lymph node dissection and esophagus reconstruction with gastric tube. Patients had an average age of 59.9 years (range 48 to 82). They were divided into two groups according to their response to nCRT; Complete response group (n = 13) and non-complete response group (n = 19) (Table 2). Four patients in the non-complete response group and two patients in the complete response group did not undergo surgery after nCRT. The response status of these patients was confirmed by clinical evaluation and endoscopic biopsy.
[Figure omitted. See PDF.]
[Figure omitted. See PDF.]
RNA expressions were different between complete response and non-complete response groups
RNA sequencing reads were mapped against the human genome assembly (Ensembl Build 37) using TopHat (v2.1.1). We identified 464 differentially expressed genes (fold change >2 or <2, and a DESeq p-value of < 0.05). In non-complete group, 240 genes were up-regulated, and 224 genes were down-regulated. The fold changes of top 20 up-regulated genes and top 20 down-regulated genes are presented in Table 3 and S1 Table. Unsupervised hierarchical clustering of 20 up-regulated and 20 down-regulated genes also revealed differences in complete response or non-complete response of nCRT (Fig 1).
[Figure omitted. See PDF.]
[Figure omitted. See PDF.]
To explore potential functions of the differentially expressed genes and their controlled biological processes, we used the Database for Annotation, Visualization, and Integrated Discovery (DAVID) [28, 29]. Up-regulated genes in esophageal cancer with non-complete response were grouped 60 clusters, among which 30 clusters had P-values <0.05. These genes are associated with the cellular protein metabolism process, glucose homeostasis, TGF-beta receptor signal response pathway, cholesterol homeostasis, cell differentiation and response to the drug (Fig 2A and S2 Table). The results of the analysis using Metascape are shown in Fig 2B. The up-regulated gene were mainly associated with 6 GO Biological Processes, including Orexin receptor pathway, regulation of vasculature development, response to peptide, lung development, regulation of protein kinase activity and response to bacterium. In addition, we analyzed down-regulated genes in esophageal cancer with non-complete response (S2 Fig). These genes are associated with extracellular matrix disassembly, collagen catabolic process, angiogenesis, immune response, cell adhesion and extracellular matrix organization (S2A Fig). The down-regulated gene were mainly associated with 9 GO Biological Processes, including NABA core matrisome, KRAS.DF.V1 up, PID integrin3 pathway, NABA ECM Glycoproteins, supramolecular fiber organization, BMI1 DN MEL18 DN.V1 up, wound heading, morphogenesis of an epithelium and positive regulation of cellular component biogenesis (S2B Fig).
[Figure omitted. See PDF.]
We first compared between complete response and non-complete response groups, expressions of up-regulated genes. We then selected 4 genes that are associated with cell proliferation and cell migration: namely, AGR2 [30], PPP1R15A [31], GADD45B [32], and LRG1 [33]. RNA sequencing indicated that the 4 genes were significantly up-regulated in patients with non-complete response to nCRT compared with those with complete response (Fig 3). To determine their possible risks for therapy progression in esophageal cancer patients, we plotted receiver operating characteristic curves (ROC) using read counts of RNA-Seq and therapy response of nCRT. Notably, the areas under the curves (AUCs) of AGR2, GADD45B, PPP1R15A and LRG1 genes were 0.671, 0.529, 0.483 and 0.521, respectively (Fig 4). Results suggest that AGR2 gene was associated with therapy response of nCRT in esophageal cancer patients.
[Figure omitted. See PDF.]
[Figure omitted. See PDF.]
Knockdown of AGR2 in esophageal cancer cells were more sensitive to the cytotoxicity effect of cisplatin and 5-fluorouracil
Previous studies reported that AGR2 is involved in head and neck squamous cell carcinoma by regulating cell transformation and epithelial-mesenchymal transition (EMT) signaling pathways [34], and that it also promotes tumor growth in esophageal adenocarcinoma [35]. In this study, we found that the AGR2 mRNA p-regulated in patients with non-complete response before nCRT. Therefore, we selected AGR2 for further investigation. We applied the siRNA approach to knockdown AGR2 expression in cell lines of esophageal cancer (CE146T/VGH, TE2, and CE48T/VGH) and then performed the MTT assay. Western blot analysis showed that protein levels of AGR2 were significantly reduced in CE146T/VGH, TE2, and CE48T/VGH cells transfected with si-AGR2. (S1 Fig). MTT assay showed a lower cell viability of the AGR2-knockdown esophageal cell line following treatment with 2.5 μM cisplatin and 3 μM 5-FU, while those cell viabilities of CE48T/VGH and CE146T/VGH remained unchanged (Fig 5). Treatment with 6 μM cisplatin and 20 μM 5-FU on AGR2-knockdown cells (CE48T/VGH, CE146T/VGH and TE-2) led to a lower cell viability relative to the siRNA-control (Fig 5). These finding indicated that AGR2 down-regulated cells were more sensitive to cisplatin and 5-FU combined treatment.
[Figure omitted. See PDF.]
Cisplatin and 5-FU induce apoptosis in AGR2-knockdown esophageal cancer cells
To determine if AGR2 modulates the sensitivity of esophageal cells to cisplatin and 5-FU, knockdown of AGR2 in TE2 cells were incubated with 2.5 μM cisplatin and 3 μM 5-FU for 72hr. We then assessed in vitro effects on apoptosis induction using the Annexin V (Fig 6). We found on change in the percentage of apoptosis cells in the si-AGR2 compared with si-control (5.4 ± 0.2% vs. 8.0 ± 1.3%). Of particular note, joint application of cisplatin and 5-FU on si-ARG2 TE2 cells induced more apoptosis compared with the si-control (13.47± 1.3% vs. 16.6± 0.7%) (Fig 6). Results suggested that the cytotoxicity sensitivity to cisplatin and 5-FU in esophageal cancer was associated with AGR2 expression.
[Figure omitted. See PDF.]
Discussion
In this study, we have identified a biomarker to predict nCRT responses in esophageal cancer. Even though our specimens in ESCC patients were tiny and obtained only through gastroscopic biopsy before nCRT, we had found 464 differentially expressed genes that were associated with the response to nCRT. Among them, we further found that the expression of the anterior gradient gene, AGR2, was associated with the cytotoxicity of drug response from experiments on esophageal cell lines.
Anterior gradient genes were first found in Xenopus laevis. In humans, anterior gradient proteins are distributed mostly in endoderm-derived organs, such as the lungs, stomach, small intestine, colon, and prostate [30, 36]. AGR2 was initially found in human breast cancer specimens [30], and it is a member of the disulfide isomerase family of endoplasmic reticulum (ER) proteins that catalyze protein folding and thiol-disulfide interchange reactions [37]. Derepression of AGR2 was not only found in breast cancer cells [30, 38], but also in other common adenocarcinomas, including those derived from the esophagus [39], stomach [40], lungs [41], pancreas [42], ovaries [43], and prostate [44].
AGR2 in both breast cancer and prostate cancer is likely may be associated with endocrine status and treatment response. As an ER-localized molecular chaperone, AGR2 regulates the folding, trafficking, and assembly of cysteine-rich transmembrane receptors and the cysteine-rich intestinal glycoprotein mucin [45]. In the prostate carcinoma, AGR2 is induced by androgens [46].
In terms of protein function, AGR2 is involved in cell migration, and cellular transformation, and metastasis as well as being a p53 inhibitor [37, 45, 47]. The role of AGR2 has been implicated in inflammatory bowel disease and cancer progression [48]. Pohler et al., [49] reported that AGR2 promotes colony formation in lung cancer cells (H1299), while overexpressing AGR2 in undamaged cells does not change their cell-cycle parameters. Furthermore, in prostate cancer, extracellular AGR2 combines with vascular endothelial growth factor (VEGF), before activating VEGF receptor signalling and inducing angiogenesis. Intracellular AGR2 induces EMT gene transcription through stabilizing p65, and then facilitates metastatic processes [50]. Lucia et al., [51] showed that the function of AGR2 is reduced by TGF-β and maintains the epithelial phenotype by preventing the activation of key factors involved in the process of EMT in breast cancer. The Orexin receptor type 1 (Ox1R) had pro-apoptotic properties in esophageal cancer [52].
The condition of Barrett’s esophagus is known to precede esophageal adenocarcinoma. AGR2 is universally overexpressed in the epithelium of Barrett’s esophagus and esophageal adenocarcinoma [39, 49]. In esophageal adenocarcinoma, AGR2 expression also promotes tumor growth, cell migration, and cellular transformation [35]. Dong et al., further demonstrated that AGR2 in esophageal adenocarcinoma promotes tumor growth by inducing AGR2 expression of and regulates the Hippo signaling pathway co-activator [53].
Most researchers focused their studies on the relationship between AGRs and adenocarcinoma. However, very few of them considered AGR2 roles in squamous cell carcinoma. Ma et al., [34] reported that in head and neck squamous cell carcinoma, AGR2 expression is associated with tumor grade and tumor size. They also showed that radiotherapy and chemotherapy likely induce AGR2 expression. AGR2 expression may function as a survival factor and is remarkably associated with survivin, cyclin D1, ALDH1, Sox2, Oct4, and Slung. AGR2 may affect cell apoptosis, invasion, proliferation, metastasis, and the EMT signaling pathway in squamous cell carcinoma [34, 54, 55].
Although AGR2 levels correlate with nCRT response in ESCC, the underlying mechanism remain unclear. The p53 tumor suppressor gene is involved in the regulation of the cell cycle, apoptosis, and DNA repair. However, p53 has a high mutation frequency and its exact role on the prognosis of esophageal carcinoma remains debatable [56]. On the other hand, p21, which is a cell cycle regulator appears to be a more reliable marker for predicting nCRT responses in esophageal carcinoma. Furthermore, p21 is involved in multiple pathways that are independent of p53 [56, 57]. Therefore, we postulate a possible relationship between AGR2 and p21 expressions in ESCC.
AGR2 expression is known to be linked with drug resistance. In breast carcinoma, AGR2 expression in ER a-positive patients is associated with drug resistance to tamoxifen [58]. In lung cancer, AGR2 can modulate EGFR-TKI resistance in EGFR-mutant non-small cell carcinoma [59]. In prostate adenocarcinoma, AGR2 could enhance the antitumor effect of bevacizumab [50]. In pancreatic carcinoma, AGR2 expression is related to the response to gemcitabine [60], and in an animal model, blocking monoclonal antibodies against AGR2 and C4.4A resulted in the regression of tumor invasion and increased survival [61]. Therefore, the suppression of AGR2 may be a therapeutic option.
DiMaio et al., retrospectively examined 116 specimens of esophageal carcinoma. They demonstrated that the presence of diffuse AGR2 expression is highly sensitive to esophageal adenocarcinoma. However, focal expression of AGR2 was found only in 1/3 (36.59%) of ESCC specimens [62]. Because AGR2 is not universally expressed in ESCC, a predictor is of greater importance in esophageal adenocarcinoma. Valladares-Ayerbesand et al., AGR2 as a suitable candidate gene for the detection of circulating tumor cells in patients with gastrointestinal cancer, a finding that extends the clinical application of AGR2 [63].
Supporting information
S1 Table. Top 20 down-regulated expressed genes between complete response and non-complete response groups by RNA-seq.
https://doi.org/10.1371/journal.pone.0276990.s001
(DOCX)
S2 Table. Functional analysis of Up-regulated genes.
https://doi.org/10.1371/journal.pone.0276990.s002
(DOCX)
S1 Fig. Protein levels of AGR2 decreased in esophageal cancer cell lines with AGR2-knockdown.
The amounts of 25μM control siRNA (si-Control) and 25μM AGR2 siRNA (si-AGR2) were transfected into esophageal cells. The loading control was β-actin expression level. (A) CE146T/VGH, (B) TE-2 and CE48T/VGH.
https://doi.org/10.1371/journal.pone.0276990.s003
(TIF)
S2 Fig.
The bar graph showed the down-regulated gene expression pattern according to functional enrichment analysis from DAVID (A) and Metascape (B) online.
https://doi.org/10.1371/journal.pone.0276990.s004
(TIF)
Acknowledgments
The authors would like to thank the research team of Precision Medicine Center, Taichung Veterans General Hospital for technical assistance in genetic diagnosis and analysis.
Citation: Lin C-H, Chuang H-N, Hsiao T-H, Kumar VB, Hsu C-H, Huang C-Y, et al. (2022) AGR2 expression as a predictive biomarker for therapy response in esophageal squamous cell carcinoma. PLoS ONE 17(11): e0276990. https://doi.org/10.1371/journal.pone.0276990
About the Authors:
Chih-Hung Lin
Contributed equally to this work with: Chih-Hung Lin, Han-Ni Chuang
Roles: Data curation, Investigation, Writing – original draft, Writing – review & editing
Affiliations Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, Division of Thoracic Surgery, Department of Surgery, Taichung Veteran General Hospital, Taichung, Taiwan
Han-Ni Chuang
Contributed equally to this work with: Chih-Hung Lin, Han-Ni Chuang
Roles: Formal analysis, Methodology, Writing – original draft, Writing – review & editing
Affiliation: Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
Tzu-Hung Hsiao
Roles: Investigation, Resources, Supervision
Affiliations Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan, Department of Public Health, Fu Jen Catholic University, New Taipei City, Taiwan, Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, Taiwan
V. Bharath Kumar
Roles: Writing – original draft, Writing – review & editing
Affiliation: Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
Chiung-Hung Hsu
Roles: Software
Affiliation: Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
Chih-Yang Huang
Roles: Writing – review & editing
Affiliations Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan, Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
Li-Wen Lee
Roles: Methodology, Validation, Visualization
Affiliation: Division of Thoracic Surgery, Department of Surgery, Taichung Veteran General Hospital, Taichung, Taiwan
Chien-Lin Mao
Roles: Software
Affiliation: Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
Jiunn-Liang Ko
Roles: Project administration, Resources
E-mail: [email protected] (JLK); [email protected] (CPH)
Affiliations Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, Department of Medical Oncology and Chest Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
Chung-Ping Hsu
Roles: Conceptualization, Project administration, Resources
E-mail: [email protected] (JLK); [email protected] (CPH)
Affiliations Division of Thoracic Surgery, Department of Surgery, Taichung Veteran General Hospital, Taichung, Taiwan, Division of Thoracic Surgery, Department of Surgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
https://orcid.org/0000-0002-7686-4935
1. Edgren G, Adami HO, Weiderpass E, Nyren O. A global assessment of the oesophageal adenocarcinoma epidemic. Gut. 2013;62(10):1406–14. Epub 2012/08/25. pmid:22917659.
2. Bosset JF, Gignoux M, Triboulet JP, Tiret E, Mantion G, Elias D, et al. Chemoradiotherapy followed by surgery compared with surgery alone in squamous-cell cancer of the esophagus. N Engl J Med. 1997;337(3):161–7. Epub 1997/07/17. pmid:9219702.
3. Enzinger PC, Mayer RJ. Esophageal cancer. N Engl J Med. 2003;349(23):2241–52. Epub 2003/12/06. pmid:14657432.
4. Holscher AH, Bollschweiler E, Bogoevski D, Schmidt H, Semrau R, Izbicki JR. Prognostic impact of neoadjuvant chemoradiation in cT3 oesophageal cancer—A propensity score matched analysis. Eur J Cancer. 2014;50(17):2950–7. Epub 2014/10/14. pmid:25307749.
5. van Hagen P, Hulshof MC, van Lanschot JJ, Steyerberg EW, van Berge Henegouwen MI, Wijnhoven BP, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 2012;366(22):2074–84. Epub 2012/06/01. pmid:22646630.
6. Sjoquist KM, Burmeister BH, Smithers BM, Zalcberg JR, Simes RJ, Barbour A, et al. Survival after neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal carcinoma: an updated meta-analysis. Lancet Oncol. 2011;12(7):681–92. Epub 2011/06/21. pmid:21684205.
7. Urschel JD, Vasan H. A meta-analysis of randomized controlled trials that compared neoadjuvant chemoradiation and surgery to surgery alone for resectable esophageal cancer. Am J Surg. 2003;185(6):538–43. Epub 2003/06/05. pmid:12781882.
8. MacGuill M, Mulligan E, Ravi N, Rowley S, Byrne PJ, Hollywood D, et al. Clinicopathologic factors predicting complete pathological response to neoadjuvant chemoradiotherapy in esophageal cancer. Dis Esophagus. 2006;19(4):273–6. Epub 2006/07/27. pmid:16866859.
9. Alnaji RM, Du W, Gabriel E, Singla S, Attwood K, Nava H, et al. Pathologic Complete Response Is an Independent Predictor of Improved Survival Following Neoadjuvant Chemoradiation for Esophageal Adenocarcinoma. J Gastrointest Surg. 2016;20(9):1541–6. Epub 2016/06/05. pmid:27260525.
10. Soror T, Kho G, Zhao KL, Ismail M, Badakhshi H. Impact of pathological complete response following neoadjuvant chemoradiotherapy in esophageal cancer. J Thorac Dis. 2018;10(7):4069–76. Epub 2018/09/04. pmid:30174850; PubMed Central PMCID: PMC6106005.
11. Shapiro J, van Lanschot JJB, Hulshof M, van Hagen P, van Berge Henegouwen MI, Wijnhoven BPL, et al. Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol. 2015;16(9):1090–8. Epub 2015/08/10. pmid:26254683.
12. Kumagai K, Rouvelas I, Tsai JA, Mariosa D, Klevebro F, Lindblad M, et al. Meta-analysis of postoperative morbidity and perioperative mortality in patients receiving neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal and gastro-oesophageal junctional cancers. Br J Surg. 2014;101(4):321–38. Epub 2014/02/05. pmid:24493117.
13. Mariette C, Dahan L, Mornex F, Maillard E, Thomas PA, Meunier B, et al. Surgery alone versus chemoradiotherapy followed by surgery for stage I and II esophageal cancer: final analysis of randomized controlled phase III trial FFCD 9901. J Clin Oncol. 2014;32(23):2416–22. Epub 2014/07/02. pmid:24982463.
14. Bollschweiler E, Metzger R, Drebber U, Baldus S, Vallbohmer D, Kocher M, et al. Histological type of esophageal cancer might affect response to neo-adjuvant radiochemotherapy and subsequent prognosis. Ann Oncol. 2009;20(2):231–8. Epub 2008/10/07. pmid:18836090.
15. Schneider PM, Baldus SE, Metzger R, Kocher M, Bongartz R, Bollschweiler E, et al. Histomorphologic tumor regression and lymph node metastases determine prognosis following neoadjuvant radiochemotherapy for esophageal cancer: implications for response classification. Ann Surg. 2005;242(5):684–92. Epub 2005/10/26. pmid:16244542; PubMed Central PMCID: PMC1409844.
16. Dulak AM, Stojanov P, Peng S, Lawrence MS, Fox C, Stewart C, et al. Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat Genet. 2013;45(5):478–86. Epub 2013/03/26. pmid:23525077; PubMed Central PMCID: PMC3678719.
17. Warnecke-Eberz U, Vallbohmer D, Alakus H, Kutting F, Lurje G, Bollschweiler E, et al. ERCC1 and XRCC1 gene polymorphisms predict response to neoadjuvant radiochemotherapy in esophageal cancer. J Gastrointest Surg. 2009;13(8):1411–21. Epub 2009/05/08. pmid:19421825.
18. Alakus H, Bollschweiler E, Holscher AH, Warnecke-Eberz U, Frazer KA, Harismendy O, et al. Homozygous GNAS 393C-allele carriers with locally advanced esophageal cancer fail to benefit from platinum-based preoperative chemoradiotherapy. Ann Surg Oncol. 2014;21(13):4375–82. Epub 2014/07/06. pmid:24986238.
19. Alakus H, Warnecke-Eberz U, Bollschweiler E, Monig SP, Vallbohmer D, Brabender J, et al. GNAS1 T393C polymorphism is associated with histopathological response to neoadjuvant radiochemotherapy in esophageal cancer. Pharmacogenomics J. 2009;9(3):202–7. Epub 2009/03/11. pmid:19274060.
20. Narumiya K, Metzger R, Bollschweiler E, Alakus H, Brabender J, Drebber U, et al. Impact of ABCB1 C3435T polymorphism on lymph node regression in multimodality treatment of locally advanced esophageal cancer. Pharmacogenomics. 2011;12(2):205–14. Epub 2011/02/22. pmid:21332314.
21. Ge S, Li B, Li Y, Li Z, Liu Z, Chen Z, et al. Genomic alterations in advanced gastric cancer endoscopic biopsy samples using targeted next-generation sequencing. Am J Cancer Res. 2017;7(7):1540–53. Epub 2017/07/27. pmid:28744403; PubMed Central PMCID: PMC5523034.
22. Dreyer SB, Jamieson NB, Evers L, Duthie F, Cooke S, Marshall J, et al. Feasibility and clinical utility of endoscopic ultrasound guided biopsy of pancreatic cancer for next-generation molecular profiling. Chin Clin Oncol. 2019;8(2):16. Epub 2019/05/10. pmid:31070037.
23. Wang YJ, Jiang RR, Liu HJ, Zhang B, Ye F, Bu H. [Feasibility of amplicon-based targeted next-generation sequencing of colorectal cancer in endoscopic biopsies]. Zhonghua Bing Li Xue Za Zhi. 2018;47(7):499–504. Epub 2018/07/13. pmid:29996313.
24. Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol. 2019;37(8):907–15. Epub 2019/08/04. pmid:31375807.
25. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30(7):923–30. Epub 2013/11/15. pmid:24227677.
26. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550. Epub 2014/12/18. pmid:25516281; PubMed Central PMCID: PMC4302049.
27. Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10(1):1523. Epub 20190403. pmid:30944313; PubMed Central PMCID: PMC6447622.
28. Huang da W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;37(1):1–13. Epub 2008/11/27. pmid:19033363; PubMed Central PMCID: PMC2615629.
29. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature protocols. 2009;4(1):44–57. pmid:19131956.
30. Kuang WW, Thompson DA, Hoch RV, Weigel RJ. Differential screening and suppression subtractive hybridization identified genes differentially expressed in an estrogen receptor-positive breast carcinoma cell line. Nucleic Acids Res. 1998;26(4):1116–23. Epub 1998/03/21. pmid:9461476; PubMed Central PMCID: PMC147366.
31. Wu DY, Tkachuck DC, Roberson RS, Schubach WH. The human SNF5/INI1 protein facilitates the function of the growth arrest and DNA damage-inducible protein (GADD34) and modulates GADD34-bound protein phosphatase-1 activity. J Biol Chem. 2002;277(31):27706–15. Epub 2002/05/23. pmid:12016208.
32. Jin X, Liu X, Zhang Z, Guan Y, Xv R, Li J. Identification of key pathways and genes in lung carcinogenesis. Oncol Lett. 2018;16(4):4185–92. Epub 2018/09/27. pmid:30250533; PubMed Central PMCID: PMC6144915.
33. Wang X, Abraham S, McKenzie JAG, Jeffs N, Swire M, Tripathi VB, et al. LRG1 promotes angiogenesis by modulating endothelial TGF-beta signalling. Nature. 2013;499(7458):306–11. Epub 2013/07/23. pmid:23868260; PubMed Central PMCID: PMC3836402.
34. Ma SR, Wang WM, Huang CF, Zhang WF, Sun ZJ. Anterior gradient protein 2 expression in high grade head and neck squamous cell carcinoma correlated with cancer stem cell and epithelial mesenchymal transition. Oncotarget. 2015;6(11):8807–21. Epub 2015/04/15. pmid:25871396; PubMed Central PMCID: PMC4496185.
35. Wang Z, Hao Y, Lowe AW. The adenocarcinoma-associated antigen, AGR2, promotes tumor growth, cell migration, and cellular transformation. Cancer Res. 2008;68(2):492–7. Epub 2008/01/18. pmid:18199544.
36. Sive HL, Hattori K, Weintraub H. Progressive determination during formation of the anteroposterior axis in Xenopus laevis. Cell. 1989;58(1):171–80. Epub 1989/07/14. pmid:2752418.
37. Persson S, Rosenquist M, Knoblach B, Khosravi-Far R, Sommarin M, Michalak M. Diversity of the protein disulfide isomerase family: identification of breast tumor induced Hag2 and Hag3 as novel members of the protein family. Mol Phylogenet Evol. 2005;36(3):734–40. Epub 2005/06/07. pmid:15935701.
38. Liu D, Rudland PS, Sibson DR, Platt-Higgins A, Barraclough R. Human homologue of cement gland protein, a novel metastasis inducer associated with breast carcinomas. Cancer Res. 2005;65(9):3796–805. Epub 2005/05/04. pmid:15867376.
39. Hao Y, Triadafilopoulos G, Sahbaie P, Young HS, Omary MB, Lowe AW. Gene expression profiling reveals stromal genes expressed in common between Barrett’s esophagus and adenocarcinoma. Gastroenterology. 2006;131(3):925–33. Epub 2006/09/06. pmid:16952561; PubMed Central PMCID: PMC2575112.
40. Zhang J, Jin Y, Xu S, Zheng J, Zhang QI, Wang Y, et al. AGR2 is associated with gastric cancer progression and poor survival. Oncol Lett. 2016;11(3):2075–83. Epub 2016/03/22. pmid:26998125; PubMed Central PMCID: PMC4774612.
41. Zhu H, Lam DC, Han KC, Tin VP, Suen WS, Wang E, et al. High resolution analysis of genomic aberrations by metaphase and array comparative genomic hybridization identifies candidate tumour genes in lung cancer cell lines. Cancer Lett. 2007;245(1–2):303–14. Epub 2006/03/07. pmid:16517066.
42. Lowe AW, Olsen M, Hao Y, Lee SP, Taek Lee K, Chen X, et al. Gene expression patterns in pancreatic tumors, cells and tissues. PLoS One. 2007;2(3):e323. Epub 2007/03/29. pmid:17389914; PubMed Central PMCID: PMC1824711.
43. Park K, Chung YJ, So H, Kim K, Park J, Oh M, et al. AGR2, a mucinous ovarian cancer marker, promotes cell proliferation and migration. Exp Mol Med. 2011;43(2):91–100. Epub 2011/01/05. pmid:21200134; PubMed Central PMCID: PMC3047197.
44. Kristiansen G, Pilarsky C, Wissmann C, Kaiser S, Bruemmendorf T, Roepcke S, et al. Expression profiling of microdissected matched prostate cancer samples reveals CD166/MEMD and CD24 as new prognostic markers for patient survival. J Pathol. 2005;205(3):359–76. Epub 2004/11/09. pmid:15532095.
45. Hrstka R, Podhorec J, Nenutil R, Sommerova L, Obacz J, Durech M, et al. Tamoxifen-Dependent Induction of AGR2 Is Associated with Increased Aggressiveness of Endometrial Cancer Cells. Cancer Invest. 2017;35(5):313–24. Epub 2017/04/14. pmid:28402678.
46. Bu H, Schweiger MR, Manke T, Wunderlich A, Timmermann B, Kerick M, et al. Anterior gradient 2 and 3—two prototype androgen-responsive genes transcriptionally upregulated by androgens and by oestrogens in prostate cancer cells. FEBS J. 2013;280(5):1249–66. Epub 2013/01/09. pmid:23294566.
47. Brychtova V, Vojtesek B, Hrstka R. Anterior gradient 2: a novel player in tumor cell biology. Cancer Lett. 2011;304(1):1–7. Epub 2011/03/05. pmid:21371820.
48. Brychtova V, Mohtar A, Vojtesek B, Hupp TR. Mechanisms of anterior gradient-2 regulation and function in cancer. Semin Cancer Biol. 2015;33:16–24. Epub 2015/05/06. pmid:25937245.
49. Pohler E, Craig AL, Cotton J, Lawrie L, Dillon JF, Ross P, et al. The Barrett’s antigen anterior gradient-2 silences the p53 transcriptional response to DNA damage. Mol Cell Proteomics. 2004;3(6):534–47. Epub 2004/02/18. pmid:14967811.
50. Jia M, Guo Y, Zhu D, Zhang N, Li L, Jiang J, et al. Pro-metastatic activity of AGR2 interrupts angiogenesis target bevacizumab efficiency via direct interaction with VEGFA and activation of NF-kappaB pathway. Biochim Biophys Acta Mol Basis Dis. 2018;1864(5 Pt A):1622–33. Epub 2018/02/08. pmid:29410027.
51. Sommerova L, Ondrouskova E, Vojtesek B, Hrstka R. Suppression of AGR2 in a TGF-beta-induced Smad regulatory pathway mediates epithelial-mesenchymal transition. BMC Cancer. 2017;17(1):546. Epub 20170815. pmid:28810836; PubMed Central PMCID: PMC5557473.
52. Voisin T, El Firar A, Fasseu M, Rouyer-Fessard C, Descatoire V, Walker F, et al. Aberrant expression of OX1 receptors for orexins in colon cancers and liver metastases: an openable gate to apoptosis. Cancer Res. 2011;71(9):3341–51. Epub 20110317. pmid:21415167.
53. Dong A, Gupta A, Pai RK, Tun M, Lowe AW. The human adenocarcinoma-associated gene, AGR2, induces expression of amphiregulin through Hippo pathway co-activator YAP1 activation. J Biol Chem. 2011;286(20):18301–10. Epub 2011/04/02. pmid:21454516; PubMed Central PMCID: PMC3093902.
54. Chen YT, Ho CL, Chen PK, Chen YL, Chang CF. Anterior gradient 2: a novel sensitive tumor marker for metastatic oral cancer. Cancer Lett. 2013;339(2):270–8. Epub 2013/07/10. pmid:23834814.
55. Sweeny L, Liu Z, Bush BD, Hartman Y, Zhou T, Rosenthal EL. CD147 and AGR2 expression promote cellular proliferation and metastasis of head and neck squamous cell carcinoma. Exp Cell Res. 2012;318(14):1788–98. Epub 2012/06/05. pmid:22659167; PubMed Central PMCID: PMC3951318.
56. Vallbohmer D, Lenz HJ. Predictive and prognostic molecular markers in outcome of esophageal cancer. Dis Esophagus. 2006;19(6):425–32. Epub 2006/10/31. pmid:17069584.
57. Abbas T, Dutta A. p21 in cancer: intricate networks and multiple activities. Nat Rev Cancer. 2009;9(6):400–14. Epub 2009/05/15. pmid:19440234; PubMed Central PMCID: PMC2722839.
58. Hrstka R, Nenutil R, Fourtouna A, Maslon MM, Naughton C, Langdon S, et al. The pro-metastatic protein anterior gradient-2 predicts poor prognosis in tamoxifen-treated breast cancers. Oncogene. 2010;29(34):4838–47. Epub 2010/06/10. pmid:20531310.
59. Luu TT, Bach DH, Kim D, Hu R, Park HJ, Lee SK. Overexpression of AGR2 Is Associated With Drug Resistance in Mutant Non-small Cell Lung Cancers. Anticancer Res. 2020;40(4):1855–66. Epub 2020/04/03. pmid:32234873.
60. Ramachandran V, Arumugam T, Wang H, Logsdon CD. Anterior gradient 2 is expressed and secreted during the development of pancreatic cancer and promotes cancer cell survival. Cancer Res. 2008;68(19):7811–8. Epub 2008/10/03. pmid:18829536; PubMed Central PMCID: PMC4429896.
61. Arumugam T, Deng D, Bover L, Wang H, Logsdon CD, Ramachandran V. New Blocking Antibodies against Novel AGR2-C4.4A Pathway Reduce Growth and Metastasis of Pancreatic Tumors and Increase Survival in Mice. Mol Cancer Ther. 2015;14(4):941–51. Epub 2015/02/04. pmid:25646014; PubMed Central PMCID: PMC4710371.
62. DiMaio MA, Kwok S, Montgomery KD, Lowe AW, Pai RK. Immunohistochemical panel for distinguishing esophageal adenocarcinoma from squamous cell carcinoma: a combination of p63, cytokeratin 5/6, MUC5AC, and anterior gradient homolog 2 allows optimal subtyping. Hum Pathol. 2012;43(11):1799–807. Epub 2012/07/04. pmid:22748473; PubMed Central PMCID: PMC3465493.
63. Valladares-Ayerbes M, Diaz-Prado S, Reboredo M, Medina V, Iglesias-Diaz P, Lorenzo-Patino MJ, et al. Bioinformatics approach to mRNA markers discovery for detection of circulating tumor cells in patients with gastrointestinal cancer. Cancer Detect Prev. 2008;32(3):236–50. Epub 2008/09/20. pmid:18801625.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2022 Lin et al. This is an open access article distributed under the terms of the Creative Commons Attribution License: http://creativecommons.org/licenses/by/4.0/ (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Despite multidisciplinary therapy, the prognosis is poor for esophageal squamous cell carcinoma (ESCC). In the locally advanced stage, neoadjuvant chemoradiotherapy (nCRT) followed by surgery could provide survival benefits to some patients. Here, we aimed to identify for tumor therapy response a biomarker based on RNA sequencing. We collected endoscopic biopsies of 32 ESCC patients, who were divided according to nCRT response, into two groups: the complete response group (n = 13) and the non-complete response group (n = 19). RNA-sequencing data showed that 464 genes were differentially expressed. Increased in non-complete response group, 4 genes increased expressions were AGR2 (anterior gradient 2), GADD45B (growth arrest and DNA damage inducible beta), PPP1R15A (protein phosphatase 1 regulatory subunit 15A) and LRG1 (leucine rich alpha-2-glycoprotein 1). The areas under the curve (AUC) of the AGR2 gene was 0.671 according to read counts of RNA-seq and therapy response of nCRT. In vitro study showed that apoptosis cell was significantly increased in the AGR2-knockdown TE-2 cell line treated with cisplatin and 5-Fluorouracil (5-FU), when compared with si-control. Results suggest that in ESCC, the AGR2 gene is a promising and predictive gene marker for the response to anti-tumor therapy.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer