1
|
Mesquita P, Coelho A, Ribeiro AS, Póvoas LFC, de Oliveira C, Leça N, Silva S, Ferreira D, Pádua D, Coelho R, Jerónimo C, Paredes J, Conde C, Pereira B, Almeida R. The RNA-binding protein YTHDF3 affects gastric cancer cell migration and response to paclitaxel by regulating EZRIN. Gastric Cancer 2025:10.1007/s10120-025-01620-y. [PMID: 40366509 DOI: 10.1007/s10120-025-01620-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 04/26/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Gastric cancer (GC) is the fourth most common cause of cancer-related mortality and the fifth most common cancer worldwide. Despite efforts, the identification of biomarkers and new therapeutic approaches for GC remains elusive. Recent studies have begun to reveal the role of N6-adenosine methylation (m6A) in the regulation of gene expression. METHODS The expression of the reader YT521-B homology domain-containing family 3 (YTHDF3) in GC was assessed in 331 patients using immunohistochemistry. GC cell lines depleted of YTHDF3 using CRISPR-Cas9 were evaluated for migration, metastasis, orientation of the mitotic spindle, and response to paclitaxel. The association between YTHDF3 and EZRIN (EZR) mRNA was shown using RNA sequencing, immunofluorescence, real-time PCR, and RNA immunoprecipitation. The single-base elongation- and ligation-based qPCR amplification (SELECT) method was used to map m6A in the EZR transcript. RESULTS YTHDF3 was significantly overexpressed in GC, and high levels of YTHDF3 were predictive of the response to chemotherapy. In GC cell lines, YTHDF3 was the most highly expressed reader protein. YTHDF3 depletion impaired cytoskeleton organization, cell migration and metastasis, and orientation of the mitotic spindle, leading to an increased response to paclitaxel. EZR was one of the downregulated targets in the YTHDF3 knockout cell models and was associated with the observed phenotype. CONCLUSION YTHDF3 contributes to cell motility and response to paclitaxel in GC cell lines, at least in part through EZR regulation. The YTHDF3-EZR regulatory axis is a novel molecular player in GC, with clinical relevance and potential therapeutic utility.
Collapse
Affiliation(s)
- Patrícia Mesquita
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-465, Porto, Portugal
| | - Alexandre Coelho
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
- ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal
| | - Ana S Ribeiro
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-465, Porto, Portugal
| | - Luís F C Póvoas
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
- Biology Department, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal
| | - Catarina de Oliveira
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
| | - Nelson Leça
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
- Biology Department, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal
| | - Sara Silva
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
| | - Diana Ferreira
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
| | - Diana Pádua
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
- ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal
| | - Ricardo Coelho
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, 4031, Basel, Switzerland
| | - Carmen Jerónimo
- ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072, Porto, Portugal
| | - Joana Paredes
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-465, Porto, Portugal
- Pathology Department, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - Carlos Conde
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
- ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal
- IBMC - Institute of Molecular and Cell Biology, University of Porto, 4200-135, Porto, Portugal
| | - Bruno Pereira
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-465, Porto, Portugal
| | - Raquel Almeida
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal.
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-465, Porto, Portugal.
- Biology Department, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal.
| |
Collapse
|
2
|
Qu C, Yang H. Prognostic Significance and Immune Environment Analysis Using PANoptosis Molecular Clustering in Gastric Cancer. Med Sci Monit 2025; 31:e947710. [PMID: 40317125 PMCID: PMC12057512 DOI: 10.12659/msm.947710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/27/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) is a common malignant tumor, known for its poor prognosis and challenges in early detection. PANoptosis, a recently discovered form of cell death, is characterized by the integrated activation of pyroptosis, apoptosis, and/or necroptosis pathways. The connection between PANoptosis and the initiation, progression, and prognosis of gastric cancer remains inadequately investigated. MATERIAL AND METHODS Previous research has identified 19 PANoptosis-related genes (PRGs). Using these genes, we performed an in-depth analysis of gastric cancer to identify differentially expressed genes related to prognosis (PRDEGs). These differentially expressed genes were subsequently identified. We analyzed the risk scores, prognoses, and immune landscapes of the patients. Confirmed PRGs and gene clusters have been linked to cancer initiation and progression, patient survival, and immunity. Risk scores were computed, and patients were categorized into 2 groups on the basis of prognostic characteristics linked to 8 specific genes. To increase the accuracy of predicting patient survival, we developed a nomogram that integrates the risk score with various clinical characteristics. RESULTS The analysis revealed that gastric cancer patients classified into high-risk subgroups experienced reduced survival times and a diminished response to immunotherapy. We also found that risk scores demonstrated correlations with immune cell infiltration, tumor microenvironment characteristics (TME), and cancer stem cell (CSC) levels. The differential expression of GPA33 and APOD between gastric tumor and normal tissues was validated by RT-qPCR and immunohistochemical data from the Human Protein Atlas (HPA). In conclusion, our research indicates that genes linked to PANoptosis may serve as key indicators for evaluating the prognosis and survival rates of patients with gastric cancer. CONCLUSIONS This research has the potential to improve the early detection of gastric cancer and contribute to the development of more effective therapeutic approaches.
Collapse
|
3
|
Tanabe H, Uehara T, Ota H. Cell lineage-specific immunohistochemical markers in biliary intraepithelial neoplasia: Implications for subclassification and grading. Pathol Res Pract 2025; 269:155896. [PMID: 40056752 DOI: 10.1016/j.prp.2025.155896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Biliary intraepithelial neoplasia (BilIN), a precursor to cholangiocarcinoma, is categorized into low- and high-grade based on histological characteristics. Although gastric, intestinal, and biliary phenotypes of BilIN have been identified, detailed analyses of their immunophenotypic profiles using cell lineage-specific markers remain limited. This study aimed to define the immunohistochemical profiles of BilIN lesions, subclassify them based on their immunophenotypes, and correlate these profiles with histological grades. We examined 77 BilIN lesions from extrahepatic bile ducts, including 30 low- and 47 high-grade lesions, using immunohistochemical staining for gastric (claudin-18, MUC5AC), intestinal (cadherin-17, glycoprotein A33), and biliary (carbohydrate sulfotransferase 4) markers, along with progression markers (S100P, IMP3). Expression levels were semiquantitatively scored and correlated with histopathological features. BilIN lesions were classified into four immunophenotypes: gastric (G-type), intestinal (I-type), gastrointestinal (GI-type), and biliary (B-type). Low-grade lesions comprised G- (33.3 %), GI- (40 %), I- (13.3 %), and B-types (13.3 %), while high-grade lesions included G- (40.4 %), GI- (29.8 %), I- (21.3 %), and B-types (8.5 %). In low-grade BilIN, G-type lesions corresponded to gastric mucous cells, I-type to intestinal epithelial cells, and B-type to bile duct epithelial cells, while most GI-type lesions exhibited mixed G- and I-type components. High-grade BilIN differentiation based solely on histological characteristics was challenging to delineate due to overlapping features among I-, GI-, and B-type cells. S100P and IMP3 expression levels were significantly elevated in high-grade lesions, particularly within the I+B-type BilIN group, with no notable differences in G- or GI-type BilIN. Immunophenotypic profiling with lineage-specific markers effectively subclassified BilIN, enhancing the understanding of its histogenesis and progression.
Collapse
Affiliation(s)
- Heiwa Tanabe
- Department of Health and Medical Sciences, Shinshu University Graduate School of Medicine, Matsumoto, Japan.
| | - Takeshi Uehara
- Department of Laboratory Medicine, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Hiroyoshi Ota
- Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto, Japan.
| |
Collapse
|
4
|
Mao C, Li S, Fan R, Zhang J, Fan X, Shentu Z, Zhuang Z, Gan L. Development and Characterization of the [ 177Lu]Lu-Labeled Anti-CDH17 Nanobody Derivative for Radioimmunotherapy in the Gastric Cancer Xenograft Model. Mol Pharm 2025; 22:2077-2087. [PMID: 40088168 PMCID: PMC11980787 DOI: 10.1021/acs.molpharmaceut.4c01285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
Cadherin 17 (CDH17) is highly expressed in digestive system cancers, and the potential of nanobodies targeting CDH17 as imaging probes and delivery vehicles for radioactive β-particles warrants exploration for their theranostic potential in CDH17-overexpressing gastric cancer (GC). In this study, we screened an anti-CDH17 nanobody library and constructed two antibodies: anti-CDH17 VHH (recombinant nanobody fused with a polyhistidine tag) and anti-CDH17 VHH-ABD (recombinant nanobody fused with an albumin-binding domain). VHH targeting CDH17 and its derivative VHH-ABD were conjugated with DOTA and labeled with radionuclide 177Lu. The pharmacokinetics and theranostic efficacy of these agents were evaluated in the GC xenograft models. [177Lu]Lu-VHH and [177Lu]Lu-VHH-ABD exhibited high radiochemical purity (>99%, n = 3) and successfully delineated CDH17-positive gastric cancer tissues on SPECT/CT imaging. Compared with the rapid renal clearance of [177Lu]Lu-VHH, [177Lu]Lu-VHH-ABD demonstrated prolonged circulation times with increased and sustained tumor accumulation. Survival experiments in the MKN-45 tumor model revealed that two doses of [177Lu]Lu-VHH-ABD effectively suppressed tumor growth, with limited systemic biotoxicity. Histological analysis using hematoxylin and eosin (H&E) staining and Ki67 immunohistochemistry confirmed structural disruption and low tumor cell proliferative activity in the tumor tissue. In preclinical studies, [177Lu]Lu-anti-CDH17 VHH-ABD demonstrated substantial antitumor efficacy with manageable toxicity, offering promising clinical potential as a viable therapeutic option for CDH17-overexpressing GC.
Collapse
Affiliation(s)
- Chenkai Mao
- Center for
Cancer Diagnosis and Treatment, The Second
Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215000, China
| | - Shicheng Li
- Center for
Cancer Diagnosis and Treatment, The Second
Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215000, China
| | - Rencai Fan
- Center for
Cancer Diagnosis and Treatment, The Second
Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215000, China
| | - Jiaqi Zhang
- Center for
Cancer Diagnosis and Treatment, The Second
Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215000, China
| | - Xinying Fan
- Department
of General Practice, Zhongshan Hospital
of Fudan University, Shanghai 200032, China
| | - Zhen Shentu
- Department
of Ultrasound, Shenzhen Children’s Hospital, Shenzhen Pediatrics Institute of Shantou University Medical College, No. 7019, Yitian Road, Futian District, Shenzhen 518026, P.R. China
| | - Zhixiang Zhuang
- Center for
Cancer Diagnosis and Treatment, The Second
Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215000, China
| | - Lei Gan
- Center for
Cancer Diagnosis and Treatment, The Second
Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215000, China
| |
Collapse
|
5
|
Kang SU, Nam SJ, Kwon OB, Yim I, Kim TH, Yeo NY, Lim MN, Kim WJ, Park SW. Predictive Mortality and Gastric Cancer Risk Using Clinical and Socio-Economic Data: A Nationwide Multicenter Cohort Study. Cancers (Basel) 2024; 17:30. [PMID: 39796661 PMCID: PMC11718814 DOI: 10.3390/cancers17010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: Gastric cancer is a leading cause of cancer-related mortality, particularly in East Asia, with a notable burden in Republic of Korea. This study aimed to construct and develop machine learning models for the prediction of gastric cancer mortality and the identification of risk factors. Methods: All data were acquired from the Korean Clinical Data Utilization for Research Excellence by multiple medical centers in South Korea. A total of 23,717 gastric cancer patients were divided into two groups by cause of mortality (all-cause of 2664 and disease-specific of 1620) and investigated. We used comprehensive data integrating clinical, pathological, lifestyle, and socio-economic factors. Cox proportional hazards analysis was conducted to estimate hazard ratios for mortality. Five machine learning models (random forest, gradient boosting machine, XGBoost, light GBM, and cat boosting) were developed to predict mortality. The models were interpreted by SHAP, one of the explainable AI techniques. Results: For all-cause mortality, the gradient-boosting machine learning model demonstrated the highest performance with an AUC-ROC of 0.795. For disease-specific mortality, the light GBM model outperformed others, achieving an AUC-ROC of 0.867. Significant predictors included the AJCC7 stage, tumor size, lymph node count, and lifestyle factors such as smoking, drinking, and diabetes. Conclusions: This study underscores the importance of integrating both clinical and lifestyle data to enhance mortality prediction accuracy in gastric cancer patients. The findings highlight the need for personalized treatment approaches in the Korean population and emphasize the role of demographic-specific data in predictive modeling.
Collapse
Affiliation(s)
- Seong Uk Kang
- Department of Bigdata, Kangwon National University Hospital, Chuncheon 24289, Republic of Korea;
- Department of Convergence Security, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Seung-Joo Nam
- Department of Gastroenterology, Kangwon National University Hospital, Chuncheon 24289, Republic of Korea;
| | - Oh Beom Kwon
- Department of Pulmonology, Kangwon National University Hospital, Chuncheon 24289, Republic of Korea;
| | - Inhyeok Yim
- Department of Family Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Republic of Korea
| | - Tae-Hoon Kim
- University-Industry Cooperation Foundation, Kangwon National University, Chuncheon 24341, Republic of Korea;
| | - Na Young Yeo
- Department of Medical Big-Data Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea;
| | - Myoung Nam Lim
- Biomedical Research Institute, Kangwon National University Hospital, Chuncheon 24289, Republic of Korea;
| | - Woo Jin Kim
- Department of Internal Medicine, Kangwon National University Hospital, Chuncheon 24289, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Sang Won Park
- Department of Next Generation Information Center, Kangwon National University Hospital, Chuncheon 24289, Republic of Korea
- Department of Data Science, Weknew Co., Ltd., Chuncheon 24341, Republic of Korea
| |
Collapse
|
6
|
Yi N, Yin X, Feng X, Ren M, Ma C. Identification of gastric cancer subtypes based on disulfidptosis-related genes: GPC3 as a novel biomarker for prognosis prediction. Discov Oncol 2024; 15:810. [PMID: 39695020 DOI: 10.1007/s12672-024-01694-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
Gastric cancer (GC) is the fourth most common cancer type. "Disulfidptosis," a distinct form of cell death, is initiated through aberrant intracellular disulfide metabolism. Here, we identified various GC subtypes based on disulfidptosis-related genes (DRGs) and constructed a risk score model to identify relevant genes to help predict patient prognosis and guide treatment. We downloaded RNA sequencing (RNA-seq) data from the TCGA-STAD database, performed a difference analysis, and combined the data with GSE84437 to successfully perform an unsupervised clustering analysis based on DRGs and differentially expressed genes (DEGs). Risk-scoring models were established by screening prognosis-related DEGs. The GC samples were segregated into high-risk (HR) and low-risk (LR) groups according to their risk scores. We then evaluated the genes screened with the model in terms of prognosis, tumor, and immune cell infiltration. The response of patients with GC to immunological therapy was assessed using tumor mutational burden, microsatellite instability, and tumor immune dysfunction and exclusion scores. Using unsupervised cluster analysis, we identified two DRG clusters and two gene clusters that differed in prognosis and tumor microenvironment. A six-gene model was developed for risk score assessment. The LR group demonstrated superior performance compared to the HR group in terms of immunity, exhibiting greater sensitivity to immunotherapy. Thereafter, we selected the model gene GPC3 for single-gene analysis and verified it by experimental validation. The results demonstrated that GPC3 can serve as a standalone biomarker with promising clinical applicability in the prognostic prediction and clinical management of GC.
Collapse
Affiliation(s)
- Nan Yi
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Xindong Yin
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Xiao Feng
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Ming Ren
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China.
| | - Chaoqun Ma
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
7
|
Li J, Xie B, Wang H, Wang Q, Wu Y. Investigating MATN3 and ASPN as novel drivers of gastric cancer progression via EMT pathways. Hum Mol Genet 2024; 33:2035-2050. [PMID: 39301785 DOI: 10.1093/hmg/ddae129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/09/2024] [Accepted: 08/24/2024] [Indexed: 09/22/2024] Open
Abstract
Gastric cancer (GC) is a leading cause of cancer-related deaths globally, necessitating the identification of novel therapeutic targets. This study investigates the roles of MATN3 and ASPN in GC progression via the epithelial-mesenchymal transition (EMT) pathway. Analysis of the Cancer Genome Atlas-Stomach Adenocarcinoma (TCGA-STAD) dataset revealed that both MATN3 and ASPN are significantly upregulated in GC tissues and correlate with poor patient survival. Protein-protein interaction and co-expression analyses confirmed a direct interaction between MATN3 and ASPN, suggesting their synergistic role in EMT activation. Functional assays demonstrated that MATN3 promotes GC cell proliferation, migration, and invasion, while its knockdown inhibits these malignant behaviors and induces apoptosis. ASPN overexpression further amplified these oncogenic effects. In vivo, studies in a mouse model corroborated that co-overexpression of MATN3 and ASPN enhances tumor growth and metastasis. These findings highlight the MATN3-ASPN axis as a potential therapeutic target in GC, offering new insights into the molecular mechanisms driving GC progression.
Collapse
Affiliation(s)
- Jing Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou 215004, Jiangsu Province, China
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - Bo Xie
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - Hu Wang
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - QingKang Wang
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu 233004, Anhui Province, China
| | - YongYou Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou 215004, Jiangsu Province, China
| |
Collapse
|
8
|
Vilarinho T, Pádua D, Pereira B, Mesquita P, Almeida R. MISP Is Overexpressed in Intestinal Metaplasia and Gastric Cancer. Curr Oncol 2024; 31:2769-2779. [PMID: 38785491 PMCID: PMC11120023 DOI: 10.3390/curroncol31050210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Gastric cancer is the fifth most common cancer and the fourth cause of global cancer mortality. The identification of new biomarkers and drug targets is crucial to allow the better prognosis and treatment of patients. The mitotic spindle positioning (MISP) protein has the function of correcting mitotic spindle positioning and centrosome clustering and has been implicated in the cytokinesis and migration of cancer cells. The goal of this work was to evaluate the expression and clinical relevance of MISP in gastric cancer. MISP expression was evaluated by immunohistochemistry in a single hospital series (n = 286) of gastric adenocarcinomas and compared with normal gastric mucosa and intestinal metaplasia, a preneoplastic lesion. MISP was detected on the membrane in 83% of the cases, being overexpressed in gastric cancer compared to normal gastric mucosa (n = 10). Its expression was negatively associated with diffuse and poorly cohesive types. On the other hand, it was strongly expressed in intestinal metaplasia where it was associated with MUC2 and CDX2 expression. Furthermore, when we silenced MISP in vitro, a significant decrease in the viability of gastric carcinoma cells was observed. In conclusion, MISP is overexpressed in gastric cancer, being associated with an intestinal phenotype in gastric carcinogenesis and having a role in cellular proliferation.
Collapse
Affiliation(s)
- Tomás Vilarinho
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (T.V.); (D.P.); (B.P.); (P.M.)
| | - Diana Pádua
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (T.V.); (D.P.); (B.P.); (P.M.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-465 Porto, Portugal
| | - Bruno Pereira
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (T.V.); (D.P.); (B.P.); (P.M.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-465 Porto, Portugal
| | - Patrícia Mesquita
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (T.V.); (D.P.); (B.P.); (P.M.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-465 Porto, Portugal
| | - Raquel Almeida
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (T.V.); (D.P.); (B.P.); (P.M.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-465 Porto, Portugal
- Biology Department, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| |
Collapse
|
9
|
Gao X, Han W, Chen L, Li H, Zhou F, Bai B, Yan J, Guo Y, Liu K, Li W, Li R, Yuan Q, Zhang J, Lu Y, Zhao X, Ji G, Li M, Zhao Q, Wu K, Li Z, Nie Y. Association of CDX2 and mucin expression with chemotherapeutic benefits in patients with stage II/III gastric cancer. Cancer Med 2023; 12:17613-17631. [PMID: 37602699 PMCID: PMC10523976 DOI: 10.1002/cam4.6379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
BACKGROUND Better predictors of patients with stage II/III gastric cancer (GC) most likely to benefit from adjuvant chemotherapy are urgently needed. This study aimed to assess the ability of CDX2 and mucin markers to predict prognosis and fluorouracil-based adjuvant chemotherapy benefits. METHODS CDX2 and mucin protein expressions were examined by immunohistochemistry and compared with survival and adjuvant chemotherapy benefits in a prospective evaluation cohort of 782 stage II/III GC patients. Then, the main findings were validated in an independent validation cohort (n = 386) and an external mRNA sequencing dataset (ACRG cohort, n = 193). RESULTS In the evaluation cohort, CDX2, CD10, MUC2, MUC5AC, and MUC6 expressions were observed in 59.7%, 26.7%, 27.6%, 55.1%, and 57.7% of patients, respectively. However, only the expression of CDX2 was found to be associated with adjuvant chemotherapy benefits. Most importantly, CDX2-negative patients had a poorer prognosis when treated with surgery only, while the prognosis of CDX2-negative and CDX2-positive patients was similar when receiving postoperative adjuvant chemotherapy. Further analysis revealed that patients with CDX2 negative tumors benefited from chemotherapy (5-year overall survival rates: 60.0% with chemotherapy vs. 23.2% with surgery-only, p < 0.001), whereas patients with CDX2 positive tumors did not (pinteraction = 0.004). Consistent results were obtained in the validation and ACRG cohorts. CONCLUSIONS Negative expression of CDX2 is an independent risk factor for survival in stage II/III GC, but subsequent adjuvant chemotherapy is able to compensate for this unfavorable effect. Therefore, active chemotherapy is more urgent for patients with negative CDX2 expression than for patients with positive CDX2 expression.
Collapse
Affiliation(s)
- Xianchun Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
- Department of Health Statistics, Shaanxi Key Laboratory of Free Radical Biology and Medicine and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Preventive MedicineFourth Military Medical UniversityXi'anChina
| | - Weili Han
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Ling Chen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic MedicineFourth Military Medical UniversityXi'anChina
| | - Hongwei Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Fenli Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Bin Bai
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Junya Yan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Yong Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic MedicineFourth Military Medical UniversityXi'anChina
| | - Kun Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Wenjiao Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Renlong Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Qiangqiang Yuan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Jiehao Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Yuanyuan Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Xiaodi Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Gang Ji
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Mengbin Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Qingchuan Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Kaichun Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Zengshan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic MedicineFourth Military Medical UniversityXi'anChina
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| |
Collapse
|
10
|
Wang Y, Li Y, Wang B, Ran D, Zhu C, Li P, Jiang B, Wang S. Early onset, development and histological features of gastric signet-ring cell carcinoma. Front Oncol 2023; 13:1166549. [PMID: 37483506 PMCID: PMC10361758 DOI: 10.3389/fonc.2023.1166549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/23/2023] [Indexed: 07/25/2023] Open
Abstract
Objective To explore the early onset, development and histological features of gastric signet-ring cell carcinoma (SRCC). Methods Three hundred and sixty-two patients with differentiated adenocarcinoma with signet-ring cells were enrolled. Histomorphological and immunohistochemical features and patterns of the specimens were observed in detail. Results Infection of the gastric mucosa, especially by Helicobacter pylori, can cause massive cell proliferation and transformation in the deep gastric foveola, the isthmus of the gastric gland, and the proliferative zone of the upper neck of the gland. Signet-ring-like heterocysts monoclonally proliferated after the redifferentiation and reproliferation, extending horizontally along the gastric foveola. Gastric foveolar-type SRCC grew infiltratively into the lamina propria of the mucosa and the submucosa, signet-ring cells could differentiate into undifferentiated adenocarcinoma with signet-ring cell differentiation, mucinous adenocarcinoma with signet-ring cell differentiation, gastric adenocarcinoma with signet-ring cell differentiation, and fundus gland adenocarcinoma with signet-ring cell differentiation. Conclusion Early SRCC developed from the proliferative zones of the fundus of the gastric foveola and the neck of the gastric gland, growing horizontally along the gastric foveola. It developed into gastric adenocarcinoma with signet-ring cell differentiation after reproliferation and retransformation in the mucosa.
Collapse
Affiliation(s)
- Yangkun Wang
- Department of Pathology, Shenzhen Longgang District Fourth People’s Hospital, Shenzhen, China
| | | | - Bin Wang
- Department of Radiation Therapy, Cancer Center, Shanghai Jiahui International Hospital, Shanghai, China
| | - Dongmei Ran
- Department of Pathology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Chaoya Zhu
- Department of Pathology, Third Affiliated Hospital of Zhengzhou University, Shenzhen, China
| | - Ping Li
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bo Jiang
- Department of Pathology, No. 990 Hospital of The People's Liberation Army (PLA) Joint Logistics Support Force, Zhumadian, China
| | | |
Collapse
|
11
|
Liu H, Zhang X, Fang C, Li S. Resveratrol induces the growth inhibition of CDX-deficient gastric cancer cells using CDX2 and RUNX3 via the β-catenin/TCF4 signaling pathway. Transl Oncol 2023; 35:101727. [PMID: 37354639 DOI: 10.1016/j.tranon.2023.101727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/04/2023] [Accepted: 06/17/2023] [Indexed: 06/26/2023] Open
Abstract
This study aimed to determine the expression levels of runt-related transcription factor 3 (RUNX3) and caudal-related homeobox 2 (CDX2) in patients with chronic gastritis, intestinal metaplasia, atypical hyperplasia, and gastric cancer (GC). To analyze the overexpression of CDX2 and the effects of resveratrol (Res) on MKN7 and TMK1 cells, immunohistochemical staining was performed to determine the protein expression levels in tissue samples. The biological activity of MKN7 and TMK1 cells was determined. Relative mRNA and protein expression levels were also determined. RUNX3 expression was positively correlated with CDX2 expression and negatively correlated with β-catenin and transcription factor 4 (TCF-4) levels in GC tissues. Interestingly, RUNX3 expression was negatively correlated with CDX2 expression in other tissues. CDX2 overexpression or Res treatment inhibited cell proliferation, migration, and invasion, while inducing cell apoptosis. Furthermore, RUNX3 and B-cell lymphoma-2 (Bcl-2)-associated X protein (Bax) expression levels were increased, while those of of β-catenin, TCF-4, and Bcl-2 were decreased in the CDX2 group. Upon treatment with lithium chloride (LiCl), the proliferation, migration, and invasion of CDX2-overexpressing MKN7 and TMK1 cells were enhanced. Our results indicate that Res inhibits the growth of MKN7 and TMK1 cells by increasing RUNX3 and CDX2 expression levels, with the potential involvement of the β-catenin/TCF-4 signaling pathway.
Collapse
Affiliation(s)
- Hui Liu
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical College, Yantai, Shandong, China
| | - Xinxin Zhang
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical College, Yantai, Shandong, China
| | - Can Fang
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical College, Yantai, Shandong, China
| | - Shuguang Li
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical College, Yantai, Shandong, China.
| |
Collapse
|
12
|
Wang C, Wang Z, Zhao Y, Wang F. Neoadjuvant PD-1 Inhibitor Plus Apatinib and Chemotherapy Versus Apatinib Plus Chemotherapy in Treating Patients With Locally Advanced Gastric Cancer: A Prospective, Cohort Study. J Gastric Cancer 2023; 23:328-339. [PMID: 37129156 PMCID: PMC10154141 DOI: 10.5230/jgc.2023.23.e17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/23/2023] [Accepted: 01/28/2023] [Indexed: 05/03/2023] Open
Abstract
PURPOSE This study aimed to evaluate the efficacy and safety of neoadjuvant programmed cell death-1 (PD-1) inhibitors plus apatinib and chemotherapy (PAC) in patients with locally advanced gastric cancer (LAGC). MATERIALS AND METHODS Seventy-three patients with resectable LAGC were enrolled and named the PAC group (n=39) or apatinib plus chemotherapy (AC) group (n=34) based on the treatment they chose. Neoadjuvant therapy was administered in a 21-day cycle for 3 consecutive cycles, after which surgery was performed. RESULTS The PAC group exhibited a higher objective response rate than the AC group (74.4% vs. 58.8%, P=0.159). Moreover, the PAC group showed a numerically better response profile than the AC group (P=0.081). Strikingly, progression-free survival (PFS) (P=0.019) and overall survival (OS) (P=0.049) were prolonged, whereas disease-free survival (DFS) tended to be longer in the PAC group than in the AC group (P=0.056). Briefly, the 3-year PFS, DFS, and OS rates were 76.1%, 76.1%, and 86.7% in the PAC group and 46.9%, 49.9%, and 70.3% in the AC group, respectively. Furthermore, PAC (vs. AC) treatment (hazard ratio=0.286, P=0.034) was independently associated with prolonged PFS in multivariate Cox regression analyses. The incidence of adverse events did not differ between the two groups (all P>0.05), where leukopenia, anemia, hypertension, and other adverse events were commonly observed in the PAC group. CONCLUSIONS Neoadjuvant PAC therapy may achieve a preferable pathological response, delayed progression, and prolonged survival compared to AC therapy with a similar safety profile in patients with LAGC; however, further validation is warranted.
Collapse
Affiliation(s)
- Chunjing Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhen Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Zhao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fujing Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
13
|
Rashid G, Khan NA, Elsori D, Rehman A, Tanzeelah, Ahmad H, Maryam H, Rais A, Usmani MS, Babker AM, Kamal MA, Hafez W. Non-steroidal anti-inflammatory drugs and biomarkers: A new paradigm in colorectal cancer. Front Med (Lausanne) 2023; 10:1130710. [PMID: 36950511 PMCID: PMC10025514 DOI: 10.3389/fmed.2023.1130710] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/08/2023] [Indexed: 03/08/2023] Open
Abstract
Colorectal cancer is a sporadic, hereditary, or familial based disease in its origin, caused due to diverse set of mutations in large intestinal epithelial cells. Colorectal cancer (CRC) is a common and deadly disease that accounts for the 4th worldwide highly variable malignancy. For the early detection of CRC, the most common predictive biomarker found endogenously are KRAS and ctDNA/cfDNA along with SEPT9 methylated DNA. Early detection and screening for CRC are necessary and multiple methods can be employed to screen and perform early diagnosis of CRC. Colonoscopy, an invasive method is most prevalent for diagnosing CRC or confirming the positive result as compared to other screening methods whereas several non-invasive techniques such as molecular analysis of breath, urine, blood, and stool can also be performed for early detection. Interestingly, widely used medicines known as non-steroidal anti-inflammatory drugs (NSAIDs) to reduce pain and inflammation have reported chemopreventive impact on gastrointestinal malignancies, especially CRC in several epidemiological and preclinical types of research. NSAID acts by inhibiting two cyclooxygenase enzymes, thereby preventing the synthesis of prostaglandins (PGs) and causing NSAID-induced apoptosis and growth inhibition in CRC cells. This review paper majorly focuses on the diversity of natural and synthetic biomarkers and various techniques for the early detection of CRC. An approach toward current advancement in CRC detection techniques and the role of NSAIDs in CRC chemoprevention has been explored systematically. Several prominent governing mechanisms of the anti-cancer effects of NSAIDs and their synergistic effect with statins for an effective chemopreventive measure have also been discussed in this review paper.
Collapse
Affiliation(s)
- Gowhar Rashid
- Department of Amity Medical School, Amity University, Gurugram, India
| | - Nihad Ashraf Khan
- Department of Biosciences, Jamia Millia Islamia, Central University, New Delhi, India
| | - Deena Elsori
- Faculty of Resillience, Deans Office Rabdan Academy, Abu Dhabi, United Arab Emirates
| | - Andleeb Rehman
- Department of Biotechnology, Shri Mata Vaishno Devi University, Katra, India
| | - Tanzeelah
- Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Haleema Ahmad
- Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh, India
| | - Humaira Maryam
- Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh, India
| | - Amaan Rais
- Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh, India
| | - Mohd Salik Usmani
- The Department of Surgery, Faculty of Medicine, JNMCH, AMU, Uttar Pradesh, India
| | - Asaad Ma Babker
- Department of Medical Laboratory Sciences, Gulf Medical University, Ajman, United Arab Emirates
| | - Mohammad Azhar Kamal
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Wael Hafez
- Department of Internal Medicine, NMC Royal Hospital, Abu Dhabi, United Arab Emirates
- The Medical Research Division, Department of Internal Medicine, The National Research Center, Ad Doqi, Egypt
| |
Collapse
|
14
|
Li HS, Chu CL. Intestinal metaplasia in progression of Barrett's esophagus to esophageal adenocarcinoma. Shijie Huaren Xiaohua Zazhi 2023; 31:41-47. [DOI: 10.11569/wcjd.v31.i2.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The incidence of esophageal adenocarcinoma (EAC) has been increasing year by year. The prognosis of EAC is poor, and the 5-year survival rate is less than 20%. Barrett's esophagus (BE) is the only known precancerous lesion of EAC. BE with intestinal metaplasia (IM) has a higher risk of progressing to EAC. Exploring the mechanism of IM and finding targeted therapeutic targets for BE has become an important measure for tumor prevention. Bile acid reflux is considered an important factor in the occurrence of IM and promotes the progression of BE to EAC. However, the molecular regulatory mechanism of bile reflux induced IM and carcinogenesis remains unclear. This article reviews the environment, significance, and cell origin theory of IM, toxic effects of bile reflux, and molecular changes of IM progression to tumor, aiming to improve clinicians' understanding of IM in BE and provide evidence for early intervention of BE and prevention and treatment of EAC.
Collapse
Affiliation(s)
- Hai-Su Li
- Jinan Central Hospital, Jinan Key Translational Gastroenterology Laboratory, Jinan Digestive Diseases Clinical Research Center, Jinan 250013, Shandong Province, China
| | - Chuan-Lian Chu
- Jinan Central Hospital, Jinan Key Translational Gastroenterology Laboratory, Jinan Digestive Diseases Clinical Research Center, Jinan 250013, Shandong Province, China
| |
Collapse
|
15
|
Ma J, Xu X, Fu C, Xia P, Tian M, Zheng L, Chen K, Liu X, Li Y, Yu L, Zhu Q, Yu Y, Fan R, Jiang H, Li Z, Yang C, Xu C, Long Y, Wang J, Li Z. CDH17 nanobodies facilitate rapid imaging of gastric cancer and efficient delivery of immunotoxin. Biomater Res 2022; 26:64. [PMID: 36435809 PMCID: PMC9701387 DOI: 10.1186/s40824-022-00312-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/27/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND It is highly desirable to develop new therapeutic strategies for gastric cancer given the low survival rate despite improvement in the past decades. Cadherin 17 (CDH17) is a membrane protein highly expressed in cancers of digestive system. Nanobody represents a novel antibody format for cancer targeted imaging and drug delivery. Nanobody targeting CHD17 as an imaging probe and a delivery vehicle of toxin remains to be explored for its theragnostic potential in gastric cancer. METHODS Naïve nanobody phage library was screened against CDH17 Domain 1-3 and identified nanobodies were extensively characterized with various assays. Nanobodies labeled with imaging probe were tested in vitro and in vivo for gastric cancer detection. A CDH17 Nanobody fused with toxin PE38 was evaluated for gastric cancer inhibition in vitro and in vivo. RESULTS Two nanobodies (A1 and E8) against human CDH17 with high affinity and high specificity were successfully obtained. These nanobodies could specifically bind to CDH17 protein and CDH17-positive gastric cancer cells. E8 nanobody as a lead was extensively determined for tumor imaging and drug delivery. It could efficiently co-localize with CDH17-positive gastric cancer cells in zebrafish embryos and rapidly visualize the tumor mass in mice within 3 h when conjugated with imaging dyes. E8 nanobody fused with toxin PE38 showed excellent anti-tumor effect and remarkably improved the mice survival in cell-derived (CDX) and patient-derived xenograft (PDX) models. The immunotoxin also enhanced the anti-tumor effect of clinical drug 5-Fluorouracil. CONCLUSIONS The study presents a novel imaging and drug delivery strategy by targeting CDH17. CDH17 nanobody-based immunotoxin is potentially a promising therapeutic modality for clinical translation against gastric cancer.
Collapse
Affiliation(s)
- Jingbo Ma
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China.,College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, P. R. China
| | - Xiaolong Xu
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Chunjin Fu
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Peng Xia
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China.,Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430072, Hubei, P. R. China
| | - Ming Tian
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China.,Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430072, Hubei, P. R. China
| | - Liuhai Zheng
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Kun Chen
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Xiaolian Liu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, P. R. China
| | - Yilei Li
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, P. R. China
| | - Le Yu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, P. R. China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, P.R. China
| | - Qinchang Zhu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, P.R. China
| | - Yangyang Yu
- Health Science Center, Shenzhen University, Shenzhen, 518055, Guangdong, P. R. China
| | - Rongrong Fan
- Deapartment of Biosciences and Nutrition, Karolinska Institute, 14157, Stockholm, Sweden
| | - Haibo Jiang
- Department of Chemistry, The University of Hong Kong, Pok Fu Lam, Hong Kong, P. R. China
| | - Zhifen Li
- School of Chemistry and Chemical Engineering, Shanxi Datong University, Xing Yun Street, Pingcheng District, Datong, 037009, Shanxi, P. R. China
| | - Chuanbin Yang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Chengchao Xu
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Ying Long
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China.
| | - Jigang Wang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China. .,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, P.R. China. .,Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China.
| | - Zhijie Li
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China. .,Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China.
| |
Collapse
|
16
|
Cao F, Hu J, Yuan H, Cao P, Cheng Y, Wang Y. Identification of pyroptosis-related subtypes, development of a prognostic model, and characterization of tumour microenvironment infiltration in gastric cancer. Front Genet 2022; 13:963565. [PMID: 35923703 PMCID: PMC9340157 DOI: 10.3389/fgene.2022.963565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
As a new programmed death mode, pyroptosis plays an indispensable role in gastric cancer (GC) and has strong immunotherapy potential, but the specific pathogenic mechanism and antitumor function remain unclear. We comprehensively analysed the overall changes of pyroptosis-related genes (PRGs) at the genomic and epigenetic levels in 886 GC patients. We identified two molecular subtypes by consensus unsupervised clustering analysis. Then, we calculated the risk score and constructed the risk model for predicting prognostic and selected nine PRGs related genes (IL18RAP, CTLA4, SLC2A3, IL1A, KRT7,PEG10, IGFBP2, GPA33, and DES) through LASSO and COX regression analyses in the training cohorts and were verified in the test cohorts. Consequently, a highly accurate nomogram for improving the clinical applicability of the risk score was constructed. Besides, we found that multi-layer PRGs alterations were correlated with patient clinicopathological features, prognosis, immune infiltration and TME characteristics. The low risk group mainly characterized by increased microsatellite hyperinstability, tumour mutational burden and immune infiltration. The group had lower stromal cell content, higher immune cell content and lower tumour purity. Moreover, risk score was positively correlated with T regulatory cells, M1 and M2 macrophages. In addition, the risk score was significantly associated with the cancer stem cell index and chemotherapeutic drug sensitivity. This study revealed the genomic, transcriptional and TME multiomics features of PRGs and deeply explored the potential role of pyroptosis in the TME, clinicopathological features and prognosis in GC. This study provides a new immune strategy and prediction model for clinical treatment and prognosis evaluation.
Collapse
Affiliation(s)
- Feng Cao
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Jingtao Hu
- Aviation Hygiene Branch, China Eastern Airlines Co,.Ltd, Anhui Branch, Hefei, China
| | - Hongtao Yuan
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Pengwei Cao
- Hepatopancreatobiliary Surgery, Department of General Surgery, The First Hospital of Anhui Medical University, Hefei, China
| | - Yunsheng Cheng
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Yunsheng Cheng, ; Yong Wang,
| | - Yong Wang
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Yunsheng Cheng, ; Yong Wang,
| |
Collapse
|
17
|
Shin HG, Yang HR, Yoon A, Lee S. Bispecific Antibody-Based Immune-Cell Engagers and Their Emerging Therapeutic Targets in Cancer Immunotherapy. Int J Mol Sci 2022; 23:5686. [PMID: 35628495 PMCID: PMC9146966 DOI: 10.3390/ijms23105686] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer is the second leading cause of death worldwide after cardiovascular diseases. Harnessing the power of immune cells is a promising strategy to improve the antitumor effect of cancer immunotherapy. Recent progress in recombinant DNA technology and antibody engineering has ushered in a new era of bispecific antibody (bsAb)-based immune-cell engagers (ICEs), including T- and natural-killer-cell engagers. Since the first approval of blinatumomab by the United States Food and Drug Administration (US FDA), various bsAb-based ICEs have been developed for the effective treatment of patients with cancer. Simultaneously, several potential therapeutic targets of bsAb-based ICEs have been identified in various cancers. Therefore, this review focused on not only highlighting the action mechanism, design and structure, and status of bsAb-based ICEs in clinical development and their approval by the US FDA for human malignancy treatment, but also on summarizing the currently known and emerging therapeutic targets in cancer. This review provides insights into practical considerations for developing next-generation ICEs.
Collapse
Affiliation(s)
- Ha Gyeong Shin
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
| | - Ha Rim Yang
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
| | - Aerin Yoon
- R&D Division, GC Biopharma, Yongin 16924, Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Korea
| |
Collapse
|
18
|
Almeida GM, Pereira C, Park JH, Lemos C, Campelos S, Gullo I, Martins D, Gonçalves G, Leitão D, Neto JL, André A, Borges C, Almeida D, Lee HJ, Kong SH, Kim WH, Carneiro F, Almeida R, Yang HK, Oliveira C. CD44v6 High Membranous Expression Is a Predictive Marker of Therapy Response in Gastric Cancer Patients. Biomedicines 2021; 9:biomedicines9091249. [PMID: 34572441 PMCID: PMC8465138 DOI: 10.3390/biomedicines9091249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 01/27/2023] Open
Abstract
In gastric cancer (GC), biomarkers that define prognosis and predict treatment response remain scarce. We hypothesized that the extent of CD44v6 membranous tumor expression could predict prognosis and therapy response in GC patients. Two GC surgical cohorts, from Portugal and South Korea (n = 964), were characterized for the extension of CD44v6 membranous immuno-expression, clinicopathological features, patient survival, and therapy response. The value of CD44v6 expression in predicting response to treatment and its impact on prognosis was determined. High CD44v6 expression was associated with invasive features (perineural invasion and depth of invasion) in both cohorts and with worse survival in the Portuguese GC cohort (HR 1.461; 95% confidence interval 1.002–2.131). Patients with high CD44v6 tumor expression benefited from conventional chemotherapy in addition to surgery (p < 0.05), particularly those with heterogeneous CD44v6-positive and -negative populations (CD44v6_3+) (p < 0.007 and p < 0.009). Our study is the first to identify CD44v6 high membranous expression as a potential predictive marker of response to conventional treatment, but it does not clarify CD44v6 prognostic value in GC. Importantly, our data support selection of GC patients with high CD44v6-expressing tumors for conventional chemotherapy in addition to surgery. These findings will allow better stratification of GC patients for treatment, potentially improving their overall survival.
Collapse
Affiliation(s)
- Gabriela M Almeida
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
| | - Carla Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Doctoral Programme in Biomedicine, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
| | - Ji-Hyeon Park
- Department of Surgery, Seoul National University Hospital, Seoul 03080, Korea; (J.-H.P.); (H.-J.L.); (S.-H.K.); (H.-K.Y.)
| | - Carolina Lemos
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- UnIGENe, IBMC—Institute for Molecular and Cell Biology, 4200-135 Porto, Portugal
- ICBAS—Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Sofia Campelos
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Department of Pathology, Ipatimup Diagnostics, Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Irene Gullo
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal
| | - Diana Martins
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Department of Biomedical Laboratory Sciences, ESTeSC—Coimbra Health School, Polytechnic Institute of Coimbra, 3046-854 Coimbra, Portugal
| | - Gilza Gonçalves
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
| | - Dina Leitão
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal
| | - João Luís Neto
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Ana André
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
| | - Clara Borges
- Medical Oncology Department, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal; (C.B.); (D.A.)
| | - Daniela Almeida
- Medical Oncology Department, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal; (C.B.); (D.A.)
| | - Hyuk-Joon Lee
- Department of Surgery, Seoul National University Hospital, Seoul 03080, Korea; (J.-H.P.); (H.-J.L.); (S.-H.K.); (H.-K.Y.)
- Cancer Research Institute, Seoul National University, Seoul 03080, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University Hospital, Seoul 03080, Korea; (J.-H.P.); (H.-J.L.); (S.-H.K.); (H.-K.Y.)
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea;
| | - Fátima Carneiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal
| | - Raquel Almeida
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Han-Kwang Yang
- Department of Surgery, Seoul National University Hospital, Seoul 03080, Korea; (J.-H.P.); (H.-J.L.); (S.-H.K.); (H.-K.Y.)
- Cancer Research Institute, Seoul National University, Seoul 03080, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Carla Oliveira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Correspondence: ; Tel.: +351-220-408-800
| |
Collapse
|
19
|
Katsukura N, Watanabe S, Shirasaki T, Hibiya S, Kano Y, Akahoshi K, Tanabe M, Kirimura S, Akashi T, Kitagawa M, Okamoto R, Watanabe M, Tsuchiya K. Intestinal phenotype is maintained by Atoh1 in the cancer region of intraductal papillary mucinous neoplasm. Cancer Sci 2020; 112:932-944. [PMID: 33275808 PMCID: PMC7894004 DOI: 10.1111/cas.14755] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/20/2020] [Accepted: 11/29/2020] [Indexed: 12/11/2022] Open
Abstract
Intraductal papillary mucinous neoplasm (IPMN) is a precancerous lesion of pancreatic cancer. Although there are 4 types of IPMN, among which intestinal-type IPMN is likely to progress into invasive cancer known as colloid carcinoma, no information regarding the involvement of the intestinal phenotype in the carcinogenesis of IPMN exists. The present study was conducted to explore how the intestinal differentiation system is maintained during the tumor progression of intestinal-type IPMN using surgical resection specimens. Results showed that Atoh1, a critical transcriptional factor for intestinal differentiation toward the secretory lineages of intestinal epithelial cells, was expressed in an invasive-grade IPMN. To determine the function of Atoh1 in pancreatic cancer, we generated a pancreatic ductal adenocarcinoma (PDAC) cell line overexpressing Atoh1. In a xenograft model, we successfully induced an IPMN phenotype in PDAC cells via Atoh1 induction. Finally, for the first time, we discovered that GPA33 is expressed in intestinal-type IPMN, thereby suggesting a novel target for cancer therapy. In conclusion, the intestinal differentiation system might be maintained during tumor progression of intestinal-type IPMN. Further analysis of the function of Atoh1 in IPMN might be useful for understanding the molecular mechanism underlying the malignant potential during the tumor progression of IPMN.
Collapse
Affiliation(s)
- Nobuhiro Katsukura
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sho Watanabe
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomoaki Shirasaki
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shuji Hibiya
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshihito Kano
- Department of Clinical Oncology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Precision Cancer Medicine, Graduate School, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keiichi Akahoshi
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Susumu Kirimura
- Department of Surgical Pathology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takumi Akashi
- Department of Surgical Pathology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masanobu Kitagawa
- Department of Comprehensive Pathology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.,Center for Stem Cell and Regenerative Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.,Advanced Research Institute, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kiichiro Tsuchiya
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
20
|
Montaño-Samaniego M, Bravo-Estupiñan DM, Méndez-Guerrero O, Alarcón-Hernández E, Ibáñez-Hernández M. Strategies for Targeting Gene Therapy in Cancer Cells With Tumor-Specific Promoters. Front Oncol 2020; 10:605380. [PMID: 33381459 PMCID: PMC7768042 DOI: 10.3389/fonc.2020.605380] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer is the second cause of death worldwide, surpassed only by cardiovascular diseases, due to the lack of early diagnosis, and high relapse rate after conventional therapies. Chemotherapy inhibits the rapid growth of cancer cells, but it also affects normal cells with fast proliferation rate. Therefore, it is imperative to develop other safe and more effective treatment strategies, such as gene therapy, in order to significantly improve the survival rate and life expectancy of patients with cancer. The aim of gene therapy is to transfect a therapeutic gene into the host cells to express itself and cause a beneficial biological effect. However, the efficacy of the proposed strategies has been insufficient for delivering the full potential of gene therapy in the clinic. The type of delivery vehicle (viral or non viral) chosen depends on the desired specificity of the gene therapy. The first gene therapy trials were performed with therapeutic genes driven by viral promoters such as the CMV promoter, which induces non-specific toxicity in normal cells and tissues, in addition to cancer cells. The use of tumor-specific promoters over-expressed in the tumor, induces specific expression of therapeutic genes in a given tumor, increasing their localized activity. Several cancer- and/or tumor-specific promoters systems have been developed to target cancer cells. This review aims to provide up-to-date information concerning targeting gene therapy with cancer- and/or tumor-specific promoters including cancer suppressor genes, suicide genes, anti-tumor angiogenesis, gene silencing, and gene-editing technology, as well as the type of delivery vehicle employed. Gene therapy can be used to complement traditional therapies to provide more effective treatments.
Collapse
Affiliation(s)
- Mariela Montaño-Samaniego
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Diana M. Bravo-Estupiñan
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Oscar Méndez-Guerrero
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Ernesto Alarcón-Hernández
- Laboratorio de Genética Molecular, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Miguel Ibáñez-Hernández
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|