1
|
Leck LYW, Abd El-Aziz YS, McKelvey KJ, Park KC, Sahni S, Lane DJR, Skoda J, Jansson PJ. Cancer stem cells: Masters of all traits. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167549. [PMID: 39454969 DOI: 10.1016/j.bbadis.2024.167549] [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: 02/05/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Cancer is a heterogeneous disease, which contributes to its rapid progression and therapeutic failure. Besides interpatient tumor heterogeneity, tumors within a single patient can present with a heterogeneous mix of genetically and phenotypically distinct subclones. These unique subclones can significantly impact the traits of cancer. With the plasticity that intratumoral heterogeneity provides, cancers can easily adapt to changes in their microenvironment and therapeutic exposure. Indeed, tumor cells dynamically shift between a more differentiated, rapidly proliferating state with limited tumorigenic potential and a cancer stem cell (CSC)-like state that resembles undifferentiated cellular precursors and is associated with high tumorigenicity. In this context, CSCs are functionally located at the apex of the tumor hierarchy, contributing to the initiation, maintenance, and progression of tumors, as they also represent the subpopulation of tumor cells most resistant to conventional anti-cancer therapies. Although the CSC model is well established, it is constantly evolving and being reshaped by advancing knowledge on the roles of CSCs in different cancer types. Here, we review the current evidence of how CSCs play a pivotal role in providing the many traits of aggressive tumors while simultaneously evading immunosurveillance and anti-cancer therapy in several cancer types. We discuss the key traits and characteristics of CSCs to provide updated insights into CSC biology and highlight its implications for therapeutic development and improved treatment of aggressive cancers.
Collapse
Affiliation(s)
- Lionel Y W Leck
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Yomna S Abd El-Aziz
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Kyung Chan Park
- Proteina Co., Ltd./Seoul National University, Seoul, South Korea
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| | - Patric J Jansson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
2
|
Houvast RD, van Duijvenvoorde M, Thijse K, de Steur WO, de Geus-Oei LF, Crobach ASLP, Burggraaf J, Vahrmeijer AL, Kuppen PJK. Selecting Targets for Molecular Imaging of Gastric Cancer: An Immunohistochemical Evaluation. Mol Diagn Ther 2025; 29:213-227. [PMID: 39541080 PMCID: PMC11860997 DOI: 10.1007/s40291-024-00755-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Tumor-targeted positron emission tomography (PET) and fluorescence-guided surgery (FGS) could address current challenges in pre- and intraoperative imaging of gastric cancer. Adequate selection of molecular imaging targets remains crucial for successful tumor visualization. This study evaluated the potential of integrin αvβ6, carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), epidermal growth factor receptor (EGFR), epithelial cell adhesion molecule (EpCAM) and human epidermal growth factor receptor-2 (HER2) for molecular imaging of primary gastric cancer, as well as lymph node and distant metastases. METHODS Expression of αvβ6, CEACAM5, EGFR, EpCAM and HER2 was determined using immunohistochemistry in human tissue specimens of primary gastric adenocarcinoma, healthy surrounding stomach, esophageal and duodenal tissue, tumor-positive and tumor-negative lymph nodes, and distant metastases, followed by quantification using the total immunostaining score (TIS). RESULTS Positive biomarker expression in primary gastric tumors was observed in 86% for αvβ6, 72% for CEACAM5, 77% for EGFR, 93% for EpCAM and 71% for HER2. Tumor expression of CEACAM5, EGFR and EpCAM was higher compared to healthy stomach tissue expression, while this was not the case for αvβ6 and HER2. Tumor-positive lymph nodes could be distinguished from tumor-negative lymph nodes, with accuracy ranging from 82 to 93% between biomarkers. CEACAM5, EGFR and EpCAM were abundantly expressed on distant metastases, with expression in 88-95% of tissue specimens. CONCLUSION Our findings show that CEACAM5, EGFR and EpCAM are promising targets for molecular imaging of primary gastric cancer, as well as visualization of both lymph node and distant metastases. Further clinical evaluation of PET and FGS tracers targeting these antigens is warranted.
Collapse
Affiliation(s)
- Ruben D Houvast
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| | | | - Kira Thijse
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Wobbe O de Steur
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiation Science & Technology, Delft University of Technology, Delft, The Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands
| | - A Stijn L P Crobach
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Centre for Human Drug Research, Leiden, The Netherlands
| | | | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
3
|
Wang W, Dai Z, Ge C, Zhou X, Zhan Y, Chen C. Comprehensive analysis of basement membrane-related genes showed that NELL2 is a new therapeutic target for colorectal cancer. Discov Oncol 2025; 16:195. [PMID: 39961875 PMCID: PMC11832829 DOI: 10.1007/s12672-025-01979-5] [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: 10/10/2024] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND The basement membrane (BM), an omnipresent extracellular matrix, plays a pivotal role as a physiological element in the process of tumor metastasis. However, given the heterogeneity of colorectal cancer (CRC), prognosis is challengingly predictive. Therefore, we aim to construct a prognostic model using BM-associated genes to assess patient prognosis and clinical drug treatment effects. METHOD The Non-negative Matrix Factorization (NMF) algorithm leverages the characteristics or categories of matrix rows and columns to achieve BMAG molecular classification and further develop a model for predicting patient prognosis. ssGSEA quantified the relatively abundance of 13 immune functionalities and 16 immune cell typologies. To predict the efficacy of immunotherapy, a comprehensive investigation was conducted on the correlations between riskScores and key factors such as TME, immune checkpoints, and MMR-related genes. The CCK8 method, plate cloning method and Cell Apoptosis Assessment were used to evaluate the ability of NELL2 to affect the proliferation. RESULT We developed a powerful riskScore to predict colorectal cancer prognosis and effectively differentiate the tumor microenvironment. In clinical practice, this riskScore can also be utilized to further assess patient prognosis, thereby facilitating personalized treatment strategies. In addition, downregulation of NELL2 expression inhibits CRC cell proliferation. CONCLUSION In summary, we constructed a novel riskScore using BMAG for predicting prognosis in patients with CRC and explored the efficacy of this riskScore in predicting patient response to clinical drug therapy. Most importantly, we have identified the oncogenic role of NELL2 in CRC. By inhibiting NELL2, we can further suppress the initiation and progression of CRC.
Collapse
Affiliation(s)
- Weiguo Wang
- Department of Interventional and Vascular Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Zhengxing Dai
- Department of Interventional and Vascular Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Chen Ge
- Department of General Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Xitian Zhou
- Department of Interventional and Vascular Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Yi Zhan
- Department of Interventional and Vascular Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Chaobo Chen
- Department of General Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China.
| |
Collapse
|
4
|
Li W, Gao H, Liu J. Identified VCAM1 as prognostic gene in gastric cancer by co-expression network analysis. Discov Oncol 2024; 15:771. [PMID: 39692880 PMCID: PMC11655750 DOI: 10.1007/s12672-024-01603-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
The diffuse gastric cancer (DGC) is a malignant tumor distinct from intestinal gastric cancer (IGC). This study aims to identify genetic variances and potential diagnostic and therapeutic approaches for diverse types of gastric cancer utilizing an extensive dataset. Data from RNA sequencing and clinical pathological details were acquired from The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) dataset. Co-expression gene modules were constructed via Weighted Gene Co-Expression Network Analysis (WGCNA), followed by deciphering gene functions and protein-protein interaction networks within significantly associated modules. In total, analysis was conducted on 56,753 genes from 247 individuals with gastric cancer. Particularly, 621 genes from the green module exhibited strong associations with the Lauren type of gastric cancer. The prominent genes in the green module showed enrichment in processes such as signal transduction, immune response, and the positive regulation of GTPase activity. Noteworthy among these, VCAM1 was identified as the central gene linked to patients' prognosis. Moreover, 72 gastric cancer specimens were collected from The First Affiliated Hospital of University of Science and Technology of China. Immunohistochemical analysis demonstrated a significantly higher expression of VCAM1 in DGC compared to IGC (p = 0.019). Furthermore, it was confirmed that VCAM1 expression serves as a prognostic indicator for patients with DGC (p = 0.002), a correlation not observed in IGC (p = 0.760). In conclusion, this study identifies VCAM1 as a promising diagnostic and prognostic factor, suggesting novel avenues for diagnostic and therapeutic approaches in gastric cancer.
Collapse
Affiliation(s)
- Wenjuan Li
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei, Anhui, China
- Breast Cancer Center, Division of Life Sciences and Medicine,The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, NO. 107, West 2nd Ring Road, Hefei, Anhui, China
- Department of Oncology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Hong Gao
- Breast Cancer Center, Division of Life Sciences and Medicine,The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, NO. 107, West 2nd Ring Road, Hefei, Anhui, China
| | - Jianjun Liu
- Breast Cancer Center, Division of Life Sciences and Medicine,The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, NO. 107, West 2nd Ring Road, Hefei, Anhui, China.
- Department of Oncology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China.
| |
Collapse
|
5
|
Xuan Y, Gao Q, Wang C, Cai D. Positive peritoneal lavage fluid cytology based on isolation by size of epithelial tumor cells indicates a high risk of peritoneal metastasis. PeerJ 2024; 12:e17602. [PMID: 38952968 PMCID: PMC11216200 DOI: 10.7717/peerj.17602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/30/2024] [Indexed: 07/03/2024] Open
Abstract
Background Peritoneal metastasis (PM) is the most prevalent type of metastasis in patients with gastric cancer (GC) and has an extremely poor prognosis. The detection of free cancer cells (FCCs) in the peritoneal cavity has been demonstrated to be one of the worst prognostic factors for GC. However, there is a lack of sensitive detection methods for FCCs in the peritoneal cavity. This study aimed to use a new peritoneal lavage fluid cytology examination to detect FCCs in patients with GC, and to explore its clinical significance on diagnosing of occult peritoneal metastasis (OPM) and prognosis. Methods Peritoneal lavage fluid from 50 patients with GC was obtained and processed via the isolation by size of epithelial tumor cells (ISET) method. Immunofluorescence and fluorescence in situ hybridization (FISH) were used to identify FCCs expressing chromosome 8 (CEP8), chromosome 17 (CEP17), and epithelial cell adhesion molecule (EpCAM). Results Using a combination of the ISET platform and immunofluorescence-FISH, the detection of FCCs was higher than that by light microscopy (24.0% vs. 2.0%). Samples were categorized into positive and negative groups, based on the expressions of CEP8, CEP17, and EpCAM. Statistically significant relationships were demonstrated between age (P = 0.029), sex (P = 0.002), lymphatic invasion (P = 0.001), pTNM stage (P = 0.001), and positivity for FCCs. After adjusting for covariates, patients with positive FCCs had lower progression-free survival than patients with negative FCCs. Conclusion The ISET platform highly enriched nucleated cells from peritoneal lavage fluid, and indicators comprising EpCAM, CEP8, and CEP17 confirmed the diagnosis of FCCs. As a potential detection method, it offers an opportunity for early intervention of OPM and an extension of patient survival.
Collapse
Affiliation(s)
- Ying Xuan
- Jiangnan University, Wuxi School of Medicine, Wuxi, China
- Affiliated Hospital of Jiangnan University, Department of Oncology, Wuxi, China
| | - Qizhong Gao
- Affiliated Hospital of Jiangnan University, Department of Oncology, Wuxi, China
| | - Chenhu Wang
- Affiliated Hospital of Jiangnan University, Department of Oncology, Wuxi, China
| | - Dongyan Cai
- Affiliated Hospital of Jiangnan University, Department of Oncology, Wuxi, China
| |
Collapse
|
6
|
Yang K, Yi T. Tumor cell stemness in gastrointestinal cancer: regulation and targeted therapy. Front Mol Biosci 2024; 10:1297611. [PMID: 38455361 PMCID: PMC10918437 DOI: 10.3389/fmolb.2023.1297611] [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: 09/20/2023] [Accepted: 11/14/2023] [Indexed: 03/09/2024] Open
Abstract
The cancer stem cells are a rare group of self-renewable cancer cells capable of the initiation, progression, metastasis and recurrence of tumors, and also a key contributor to the therapeutic resistance. Thus, understanding the molecular mechanism of tumor stemness regulation, especially in the gastrointestinal (GI) cancers, is of great importance for targeting CSC and designing novel therapeutic strategies. This review aims to elucidate current advancements in the understanding of CSC regulation, including CSC biomarkers, signaling pathways, and non-coding RNAs. We will also provide a comprehensive view on how the tumor microenvironment (TME) display an overall tumor-promoting effect, including the recruitment and impact of cancer-associated fibroblasts (CAFs), the establishment of an immunosuppressive milieu, and the induction of angiogenesis and hypoxia. Lastly, this review consolidates mainstream novel therapeutic interventions targeting CSC stemness regulation.
Collapse
Affiliation(s)
- Kangqi Yang
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tuo Yi
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Hu Y, Xie Q, Zhao J, Yang R, Qin J, Li H, Zhao Y, Du X, Shi C. Interaction between the EPHB2 receptor and EFNB1 ligand drives gastric cancer invasion and metastasis via the Wnt/β-catenin/FAK pathway. Int J Biol Macromol 2024; 258:128848. [PMID: 38114003 DOI: 10.1016/j.ijbiomac.2023.128848] [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: 08/01/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023]
Abstract
The survival benefit for patients with gastric cancer (GC) is modest due to its high transfer potential. Targeted therapy for metastasis-related genes in GC may be a viable approach, however, inhibitors specifically targeting GC are limited. In this study, GC patient-derived xenografts (PDX) with metastatic burden were established via orthotopic transplantation. PCR-Array analysis of primary and metastatic tumors revealed EPH receptor B2 (EPHB2) as the most significantly upregulated gene. The interaction between the EPHB2 receptor and its cognate-specific EFNB1 ligands was high in GC and correlated with a poor prognosis. Fc-EFNB1 treatment increased the invasion and migration abilities of GC cells and induced a high EPHB2 expression. EPHB2 knockdown in GC cells completely abolished the ephrin ligand-induced effects on invasion and migration abilities. Signal transduction analysis revealed Wnt/β-catenin and FAK as downstream signaling mediators potentially inducing the EPHB2 phenotype. In conclusion, the observed deregulation of EPHB2/EFNB1 expression in GC enhances the invasive phenotype, suggesting a potential role of EPHB2/EFNB1 compound in local tumor cell invasion and the formation of metastasis.
Collapse
Affiliation(s)
- Yaohua Hu
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Department of Pathology, Affiliated Hospital of Yan'an University, Yanan, Shaanxi 716000, China
| | - Qinghua Xie
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jumei Zhao
- School of Basic Medical Sciences, Medical College of Yan'an University, 580 Bao-Ta Street, Yanan, Shaanxi 716000, China
| | - Runze Yang
- Gansu University of traditional Chinese Medicine, Lanzhou, Gansu 730030, China
| | - Jing Qin
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hui Li
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yong Zhao
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiong Du
- Department of Pathology, Affiliated Hospital of Yan'an University, Yanan, Shaanxi 716000, China.
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
8
|
Chen C, Jung A, Yang A, Monroy I, Zhang Z, Chaurasiya S, Deshpande S, Priceman S, Fong Y, Park AK, Woo Y. Chimeric Antigen Receptor-T Cell and Oncolytic Viral Therapies for Gastric Cancer and Peritoneal Carcinomatosis of Gastric Origin: Path to Improving Combination Strategies. Cancers (Basel) 2023; 15:5661. [PMID: 38067366 PMCID: PMC10705752 DOI: 10.3390/cancers15235661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 02/12/2024] Open
Abstract
Precision immune oncology capitalizes on identifying and targeting tumor-specific antigens to enhance anti-tumor immunity and improve the treatment outcomes of solid tumors. Gastric cancer (GC) is a molecularly heterogeneous disease where monoclonal antibodies against human epidermal growth factor receptor 2 (HER2), vascular endothelial growth factor (VEGF), and programmed cell death 1 (PD-1) combined with systemic chemotherapy have improved survival in patients with unresectable or metastatic GC. However, intratumoral molecular heterogeneity, variable molecular target expression, and loss of target expression have limited antibody use and the durability of response. Often immunogenically "cold" and diffusely spread throughout the peritoneum, GC peritoneal carcinomatosis (PC) is a particularly challenging, treatment-refractory entity for current systemic strategies. More adaptable immunotherapeutic approaches, such as oncolytic viruses (OVs) and chimeric antigen receptor (CAR) T cells, have emerged as promising GC and GCPC treatments that circumvent these challenges. In this study, we provide an up-to-date review of the pre-clinical and clinical efficacy of CAR T cell therapy for key primary antigen targets and provide a translational overview of the types, modifications, and mechanisms for OVs used against GC and GCPC. Finally, we present a novel, summary-based discussion on the potential synergistic interplay between OVs and CAR T cells to treat GCPC.
Collapse
Affiliation(s)
- Courtney Chen
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Audrey Jung
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Annie Yang
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Isabel Monroy
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
| | - Zhifang Zhang
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Shyambabu Chaurasiya
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Supriya Deshpande
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Saul Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yuman Fong
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Anthony K. Park
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yanghee Woo
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
9
|
Deng M, Xu Y, Yao Y, Wang Y, Yan Q, Cheng M, Liu Y. Circular RNA hsa_circ_0051246 acts as a microRNA-375 sponge to promote the progression of gastric cancer stem cells via YAP1. PeerJ 2023; 11:e16523. [PMID: 38505381 PMCID: PMC10950207 DOI: 10.7717/peerj.16523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/05/2023] [Indexed: 03/21/2024] Open
Abstract
Background Gastric cancer (GC) stem cells play an important role in GC progression. Circular RNAs (circRNAs) act as microRNA (miRNA) sponges and inhibit the biological function of miRNAs in GC cytoplasm. MiRNAs also participate in GC progress. circ_0051246 was shown to be associated with miR-375 after analyzing GC microarray data GSE78091 and GSE83521. The oncoprotein Yes-associated protein 1 (YAP1) is targeted by miR-375 and can be inactivated via the Hippo tumor suppressor pathway. Due to insufficient research on circ_0051246, this study aimed to investigate its relationship with miR-375 and YAP1 in cancer stem cells (CSCs). Methods SGC-7901 CSCs were used to establish knockdown/overexpression models of circ_0051246, miR-375, and YAP1. Malignant phenotypes of CSCs were assessed using Cell Counting Kit 8, colony/sphere formation, 5-Ethynyl-2'-deoxyuridine assay, flow cytometry, Transwell, and wound healing assays. To detect the interactions between circ_0051246, miR-375, and YAP1 in CSCs, a dual-luciferase reporter assay and fluorescence in situ hybridization were performed. In addition, 24 BALB/c nude mice were used to establish orthotopic xenograft tumor models. Four groups of mice were injected with CSCs (1 × 106 cells/100 µL) with circ_0051246 knockdown, miR-375 overexpression, or their respective control cells, and tumor progression and gene expression were observed by hematoxylin-eosin staining, immunohistochemistry. Western blot and quantitative real-time PCR were utilized to examine protein and gene expression, respectively. Results Circ_0051246 silencing reduced viability, promoted apoptosis, and inhibited proliferation, migration and invasion of CSCs. The functional effects of miR-375 mimics were comparable to those of circ_0051246 knockdown; however, the opposite was observed after miR-375 inhibitors treatment of CSCs. Furthermore, circ_0051246-overexpression antagonized the miR-375 mimics' effects on CSCs. Additionally, YAP1 overexpression promoted CSC features, such as self-renewal, migration, and invasion, inhibited apoptosis and E-cadherin levels, and upregulated the expression of N-cadherin, vimentin, YAP1, neurogenic locus notch homolog protein 1, and jagged canonical notch ligand 1. Conversely, YAP1-silenced produced the opposite effect. Moreover, miR-375 treatment antagonized the malignant effects of YAP1 overexpression in CSCs. Importantly, circ_0051246 knockdown and miR-375 activation suppressed CSC tumorigenicity in vivo. Conclusion This study highlights the promotion of circ_0051246-miR-375-YAP1 axis activation in GC progression and provides a scientific basis for research on the molecular mechanism of CSCs.
Collapse
Affiliation(s)
- Minghui Deng
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Yefeng Xu
- Department of Oncology, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Yongwei Yao
- Department of Oncology, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Yiqing Wang
- Department of Oncology, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Qingying Yan
- Department of Oncology, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Miao Cheng
- Department of Oncology, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - YunXia Liu
- Department of Oncology, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
10
|
Sun W, Yuan Y, Chen J, Bao Q, Shang M, Sun P, Peng H. Construction and validation of a novel senescence-related risk score can help predict the prognosis and tumor microenvironment of gastric cancer patients and determine that STK40 can affect the ROS accumulation and proliferation ability of gastric cancer cells. Front Immunol 2023; 14:1259231. [PMID: 37915566 PMCID: PMC10616298 DOI: 10.3389/fimmu.2023.1259231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023] Open
Abstract
Background In recent years, significant molecules have been found in gastric cancer research. However, their precise roles in the disease's development and progression remain unclear. Given gastric cancer's heterogeneity, prognosis prediction is challenging. This study aims to assess patient prognosis and immune therapy efficacy using multiple key molecules. Method The WGCNA algorithm was employed to identify modules of genes closely related to immunity. A prognostic model was established using the Lasso-Cox method to predict patients' prognosis. Single-sample gene set enrichment analysis (ssGSEA) was conducted to quantify the relative abundance of 16 immune cell types and 13 immune functions. The relationship between risk score and TMB, MSI, immune checkpoints, and DNA repair genes was examined to predict the effectiveness of immune therapy. GO and KEGG analyses were performed to explore potential pathways and mechanisms associated with the genes of interest. Single-cell RNA sequencing was utilized to investigate the expression patterns of key genes in different cell types. Results Through the WGCNA algorithm and Lasso-Cox algorithm selected KL, SERPINE1, and STK40 as key genes for constructing the prognostic model. The SSGSEA algorithm was employed to evaluate the infiltration of immune cells and immune functions in different patients, and their association with the risk score was investigated. The high-risk group exhibited lower TMB and MSI compared to the low-risk group. MMR and immune checkpoint analysis revealed a significant correlation between the risk score and multiple molecules. Finally, we also believe that STK40 is the most critical senescence-related gene affecting the progression of gastric cancer. In vitro experiments showed that ROS accumulation and cell proliferation ability of gastric cancer cells were impaired when STK40 was knocked down. Conclusion In summary, we've constructed a prognostic model utilizing key genes for gastric cancer prognosis, while also showcasing its efficacy in predicting patient response to immunotherapy.
Collapse
Affiliation(s)
- Weijie Sun
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihang Yuan
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaying Chen
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qun Bao
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengsi Shang
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haixia Peng
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Wang T, Wang C, Wang J, Wang B. An Intrabody against B-Cell Receptor-Associated Protein 31 (BAP31) Suppresses the Glycosylation of the Epithelial Cell-Adhesion Molecule (EpCAM) via Affecting the Formation of the Sec61-Translocon-Associated Protein (TRAP) Complex. Int J Mol Sci 2023; 24:14787. [PMID: 37834237 PMCID: PMC10572819 DOI: 10.3390/ijms241914787] [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: 07/31/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
The epithelial cell-adhesion molecule (EpCAM) is hyperglycosylated in carcinoma tissue and the oncogenic function of EpCAM primarily depends on the degree of glycosylation. Inhibiting EpCAM glycosylation is expected to have an inhibitory effect on cancer. We analyzed the relationship of BAP31 with 84 kinds of tumor-associated antigens and found that BAP31 is positively correlated with the protein level of EpCAM. Triple mutations of EpCAM N76/111/198A, which are no longer modified by glycosylation, were constructed to determine whether BAP31 has an effect on the glycosylation of EpCAM. Plasmids containing different C-termini of BAP31 were constructed to identify the regions of BAP31 that affects EpCAM glycosylation. Antibodies against BAP31 (165-205) were screened from a human phage single-domain antibody library and the effect of the antibody (VH-F12) on EpCAM glycosylation and anticancer was investigated. BAP31 increases protein levels of EpCAM by promoting its glycosylation. The amino acid region from 165 to 205 in BAP31 plays an important role in regulating the glycosylation of EpCAM. The antibody VH-F12 significantly inhibited glycosylation of EpCAM which, subsequently, reduced the adhesion of gastric cancer cells, inducing cytotoxic autophagy, inhibiting the AKT-PI3K-mTOR signaling pathway, and, finally, resulting in proliferation inhibition both in vitro and in vivo. Finally, we clarified that BAP31 plays a key role in promoting N-glycosylation of EpCAM by affecting the Sec61 translocation channels. Altogether, these data implied that BAP31 regulates the N-glycosylation of EpCAM and may represent a potential therapeutic target for cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Bing Wang
- College of Life Science and Health, Northeastern University, 195 Chuangxin Road, Hunnan District, Shenyang 110819, China; (T.W.); (C.W.); (J.W.)
| |
Collapse
|
12
|
Ding P, Chen P, Ouyang J, Li Q, Li S. Clinicopathological and prognostic value of epithelial cell adhesion molecule in solid tumours: a meta-analysis. Front Oncol 2023; 13:1242231. [PMID: 37664060 PMCID: PMC10468606 DOI: 10.3389/fonc.2023.1242231] [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: 06/18/2023] [Accepted: 07/27/2023] [Indexed: 09/05/2023] Open
Abstract
Background Malignant tumors, mainly solid tumors, are a significant obstacle to the improvement of life expectancy at present. Epithelial cell adhesion molecule (EpCAM), a cancer stem cell biomarker, showed widespread expression in most normal epithelial cells and most cancers. Although the clinical significance of EpCAM in various malignant solid tumors has been studied extensively, the latent relationships between EpCAM and pathological and clinical characteristics in solid tumors and differences in the roles of EpCAM among tumors have not been clearly determined. The destination point of this study was to analyze the value of EpCAM in solid tumors in clinicopathological and prognostic dimension using a meta-analysis approach. Method and materials A comprehensive and systematic search of the researches published up to March 7th, 2022, in PubMed, EMBASE, Web of Science, Cochrane library and PMC databases was performed. The relationships between EpCAM overexpression, clinicopathological characteristics, and survival outcomes were analyzed. Pooled hazard ratios (HRs) with 95% confidence intervals (CIs) and odds ratios (ORs) were estimated as indicators of the degree of correlation. This research was registered on PROSPERO (International prospective register of systematic reviews), ID: CRD42022315070. Results In total, 57 articles and 14184 cases were included in this study. High EpCAM expression had a significant coherence with a poorer overall survival (OS) (HR: 1.30, 95% CI: 1.08-1.58, P < 0.01) and a worse disease-free survival (DFS) (HR: 1.58, 95% CI: 1.28-1.95, P < 0.01), especially of gastrointestinal tumors' OS (HR: 1.50, 95% CI: 1.15-1.95, P < 0.01), and DFS (HR: 1.84, 95% CI: 1.52-2.33, P < 0.01). The DFS of head and neck tumors (HR: 2.33, 95% CI: 1.51-3.61, P < 0.01) was also associated with the overexpression of EpCAM. There were no positive relationships between the overexpression of EpCAM and sex (RR: 1.03, 95% CI: 0.99-1.07, P = 0.141), T classification (RR: 0.93, 95% CI: 0.82-1.06, P = 0.293), lymph node metastasis (RR: 0.85, 95% CI: 0.54-1.32, P = 0.461), distant metastasis (RR: 0.97, 95% CI: 0.84-1.10, P = 0.606), vascular infiltration (RR: 1.05, 95% CI: 0.85-1.29, P = 0.611), and TNM stage (RR: 0.93, 95% CI: 0.83-1.04, P = 0.187). However, the overexpression of EpCAM exhibited a significant association with the histological grades (RR: 0.88, 95% CI: 0.80-0.97, P < 0.01). Conclusion Based on pooled HRs, the positive expression of EpCAM was totally correlated to a worse OS and DFS in solid tumors. The expression of EpCAM was related to a worse OS in gastrointestinal tumors and a worse DFS in gastrointestinal tumors and head and neck tumors. Moreover, EpCAM expression was correlated with the histological grade. The results presented pointed out that EpCAM could serve as a prognostic biomarker for gastrointestinal and head and neck tumors. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42022315070.
Collapse
Affiliation(s)
- Peiwen Ding
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Panyu Chen
- Operating Room, Sichuan University West China Hospital School of Nursing, Chengdu, China
| | - Jiqi Ouyang
- Department of Gastroenterology, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Qiang Li
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shijie Li
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
13
|
Xu BB, Zheng ED, Sun HY, Huang Y, Zheng L, Lan QL, Zhou XL, Geng XG, Wang YN, Wang XY, Yu YC. Comprehensive analysis of circular RNA-associated competing endogenous RNA networks and immune infiltration in gastric cancer. Transpl Immunol 2023; 77:101793. [PMID: 36773765 DOI: 10.1016/j.trim.2023.101793] [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: 08/02/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 02/12/2023]
Abstract
BACKGROUND Circular RNA (circRNA) has been proved to be an important regulator of gastric cancer (GC). However, the role and regulatory mechanism of circrna related competitive endogenous RNA (ceRNA) in GC have not been established. METHODS CircRNA data and clinical data were obtained from the GEO and TCGA databases. The ceRNA networks were constructed and a function enrichment analysis was completed. Additionally, correlations between hub genes expression, immune cell infiltration, and clinical phenotypes were determined. The differentially expressed circRNAs and their downstream microRNAs (miRNAs) were validated by quantitative real-time polymerase chain reaction, and the hub genes were validated by western blot analysis. The migration and invasion ability of overexpressed hsa_circ_0002504 was determined by a transwell assay. RESULTS The ceRNA network contained 2 circRNAs, 3 miRNAs, and 55 messenger RNAs (mRNAs). 323 biological processes terms, 53 cellular components terms, 51 molecular functions terms, and 4 signaling pathways were revealed by the function enrichment analysis. The GSEA analysis revealed that the hub genes were positively correlated with the axon guidance and adhesion molecules pathways. The correlation analysis revealed that overexpressed EPHA4 and KCNA1 indicated poor tissue differentiation and were associated with clinically advanced stages of GC. The in vitro experiments showed that hsa_circ_0002504 was significantly down-regulated in GC cell lines. In addition, the overexpression of hsa_circ_0002504 led to a significant downregulation of hsa-miR-615-5p and hsa-miR-767-5p, as well as an upregulation of EPHA4, KCNA1, and NCAM1. Furthermore, it suppressed the migration and invasion ability of GC cells. CONCLUSIONS Hsa_circ_0002504 is a potential diagnostic biomarker for GC. High expression of EPHA4 and KCNA1 may indicate poor prognosis.
Collapse
Affiliation(s)
- Bei-Bei Xu
- Department of Gastroenterology, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, Zhejiang, China; Soochow University, Suzhou 215000, Jiangsu, China; Department of Gastroenterology, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - En-Dian Zheng
- Department of Gastroenterology, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Hao-Yue Sun
- Department of Gastroenterology, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yi Huang
- Department of General Surgery, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou 325000, Zhejiang, China
| | - Liang Zheng
- Department of Gastroenterology, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Qiao-Li Lan
- Department of Gastroenterology, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Xiao-Lu Zhou
- Department of Gastroenterology, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - Xiao-Ge Geng
- Department of Gastroenterology, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - Ya-Nan Wang
- Zhejiang University of Technology, Hangzhou 310000, Zhejiang, China
| | - Xiu-Yan Wang
- Department of Gastroenterology, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| | - Ying-Cong Yu
- Department of Gastroenterology, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| |
Collapse
|
14
|
Pallares-Rusiñol A, Bernuz M, Moura SL, Fernández-Senac C, Rossi R, Martí M, Pividori MI. Advances in exosome analysis. Adv Clin Chem 2022; 112:69-117. [PMID: 36642486 DOI: 10.1016/bs.acc.2022.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There is growing demand for novel biomarkers that detect early stage disease as well as monitor clinical management and therapeutic strategies. Exosome analysis could provide the next advance in attaining that goal. Exosomes are membrane encapsulated biologic nanometric-sized particles of endocytic origin which are released by all cell types. Unfortunately, exosomes are exceptionally challenging to characterize with current technologies. Exosomes are between 30 and 200nm in diameter, a size that makes them out of the sensitivity range to most cell-oriented sorting or analysis platforms, i.e., traditional flow cytometers. The most common methods for targeting exosomes to date typically involve purification followed by the characterization and the specific determination of their cargo. The whole procedure is time consuming, requiring thus skilled personnel as well as laboratory facilities and benchtop instrumentation. The most relevant methodology for exosome isolation, characterization and quantification is addressed in this chapter, including the most up-to-date approaches to explore the potential usefulness of exosomes as biomarkers in liquid biopsies and in advanced nanomedicine.
Collapse
Affiliation(s)
- Arnau Pallares-Rusiñol
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Spain; Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Mireia Bernuz
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Spain; Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Silio Lima Moura
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Spain; Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Carolina Fernández-Senac
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Spain; Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Rosanna Rossi
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Spain; Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Mercè Martí
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - María Isabel Pividori
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Spain; Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
15
|
Yin H, Harrison TA, Thomas SS, Sather CL, Koehne AL, Malen RC, Reedy AM, Wurscher MA, Hsu L, Phipps AI, Zaidi SHE, Newcomb PA, Peters U, Huyghe JR. T cell-inflamed gene expression profile is associated with favorable disease-specific survival in non-hypermutated microsatellite-stable colorectal cancer patients. Cancer Med 2022; 12:6583-6593. [PMID: 36341526 PMCID: PMC10067089 DOI: 10.1002/cam4.5429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The anti-tumor immune response plays a key role in colorectal cancer (CRC) progression and survival. The T cell-inflamed gene expression profile (GEP) is a biomarker predicting response to checkpoint inhibitor immunotherapy across immunogenic cancer types, but the prognostic value in CRC is unknown. We evaluated associations with disease-specific survival, somatic mutations, and examined its differentially expressed genes and pathways among 84 sporadic CRC patients from the Seattle Colon Cancer Family Registry. METHODS Gene expression profiling was performed using Nanostring's nCounter PanCancer IO 360 panel. Somatic mutations were identified by a targeted DNA sequencing panel. RESULTS The T cell-inflamed GEP was positively associated with tumor mutation burden and microsatellite instability high (MSI-H). Higher T cell-inflamed GEP had favorable CRC-specific survival (hazard ratio [HR] per standard deviation unit = 0.50, p = 0.004) regardless of hypermutation or MSI status. Analysis of recurrently mutated genes having at least 10 mutation carriers, suggested that the T cell-inflamed GEP is positively associated with RYR1, and negatively associated with APC. However, these associations were attenuated after adjusting for hypermutation or MSI status. We also found that expression of genes RPL23, EPCAM, AREG and ITGA6, and the Wnt signaling pathway was negatively associated with the T cell-inflamed GEP, which might indicate immune-inhibitory mechanisms. CONCLUSIONS Our results show that the T cell-inflamed GEP is a prognostic biomarker in non-hypermutated microsatellite-stable CRC. This also suggests that patient stratification for immunotherapy within this CRC subgroup should be explored further. Moreover, reported immune-inhibitory gene expression signals may suggest targets for therapeutic combination with immunotherapy.
Collapse
Affiliation(s)
- Hang Yin
- Institute for Public Health Genetics, University of Washington, Seattle, Washington, USA
| | - Tabitha A Harrison
- Institute for Public Health Genetics, University of Washington, Seattle, Washington, USA.,Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Sushma S Thomas
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Cassie L Sather
- Genomics Resource, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Amanda L Koehne
- Experimental Histopathology, Shared Resource, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Rachel C Malen
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Adriana M Reedy
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Michelle A Wurscher
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA.,Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Amanda I Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Syed H E Zaidi
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Polly A Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Jeroen R Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| |
Collapse
|
16
|
Qu C, Zhang H, Cao H, Tang L, Mo H, Liu F, Zhang L, Yi Z, Long L, Yan L, Wang Z, Zhang N, Luo P, Zhang J, Liu Z, Ye W, Liu Z, Cheng Q. Tumor buster - where will the CAR-T cell therapy 'missile' go? Mol Cancer 2022; 21:201. [PMID: 36261831 PMCID: PMC9580202 DOI: 10.1186/s12943-022-01669-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell (CAR-T cell) therapy based on gene editing technology represents a significant breakthrough in personalized immunotherapy for human cancer. This strategy uses genetic modification to enable T cells to target tumor-specific antigens, attack specific cancer cells, and bypass tumor cell apoptosis avoidance mechanisms to some extent. This method has been extensively used to treat hematologic diseases, but the therapeutic effect in solid tumors is not ideal. Tumor antigen escape, treatment-related toxicity, and the immunosuppressive tumor microenvironment (TME) limit their use of it. Target selection is the most critical aspect in determining the prognosis of patients receiving this treatment. This review provides a comprehensive summary of all therapeutic targets used in the clinic or shown promising potential. We summarize CAR-T cell therapies’ clinical trials, applications, research frontiers, and limitations in treating different cancers. We also explore coping strategies when encountering sub-optimal tumor-associated antigens (TAA) or TAA loss. Moreover, the importance of CAR-T cell therapy in cancer immunotherapy is emphasized.
Collapse
Affiliation(s)
- Chunrun Qu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hui Cao
- Department of Psychiatry, The Second People's Hospital of Hunan Province, The Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China.,The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lanhua Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haoyang Mo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lifu Long
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luzhe Yan
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Nan Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,One-third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Weijie Ye
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
17
|
Song B, Cao Q, Li T, Liu Y, Sun Q, Fan S, Li X. Biomarker identification of chronic atrophic gastritis and its potential drug analysis. FRONTIERS IN GASTROENTEROLOGY 2022; 1. [DOI: 10.3389/fgstr.2022.948323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
BackgroundChronic atrophic gastritis (CAG) is the first step of gastric precancerous lesions, and the study of the pathogenesis of CAG is helpful for the prevention and treatment of gastric cancer(GC). The purpose of this study is to explore the potential biomarkers and therapeutic drugs of CAG through bioinformatics analysis.MethodsThe GSE11632 dataset was downloaded from Gene Expression Omnibus (GEO) database and the differentially expressed genes (DEGs) were obtained by using GEO2R online tool. We searched GeneCard and DisGeNET databases for genes related to CAG and used the overlapping genes as final DEGs for further functional enrichment analysis and Protein-protein Interaction (PPI) network analysis. Tissue-specific expressed genes were identified by BioGPS database. Cytoscape software was used to identify key hub genes and validated them in GSE27411 data sets. The upstream miRNAs of hub gene was predicted by TargetScan, miRDB and miRWalk. Finally, run the Connectivity Map (CMap) to identify new potential drugs for the treatment of CAG.ResultsA total of 430 differentially expressed mRNA were identified in this study, including 315 up-regulated genes and 115 down-regulated genes. After intersecting with CAG-related genes in GeneCard and DisGeNET databases, 42 DEGs were obtained. 24 DEGs were identified as tissue-specific expressed genes, most of which were expressed in stomach. GO and KEGG pathway analysis showed that DGEs was mainly enriched in digestion, IL-1 production, gastric acid secretion and so on. A total of 6 hub genes were generated by cytoHubba plug-in, among which ATP4A, CFTR and EPCAM had high diagnostic value. A total of 13 overlapping miRNA were predicted by 6 hub genes.ConclusionATP4A, CFTR and EPCAM may be potential biomarkers of CAG. hsa-miR-185-5p-CFTR, hsa-miR-4644-CFTR and hsa-miR-4505-CFTR are potential RNA regulatory pathways to control the progression of CAG disease. Finally, amonafide, etoposide, mycophenolate-mofetil, cycloheximide and Emetine may be potential therapeutic drugs for CAG.
Collapse
|
18
|
Xia D, Liu J, Yong J, Li X, Ji W, Zhao Z, Wang X, Xiao C, Wu S, Liu H, Zhao H, He Y. Strategies for understanding the role of cellular heterogeneity in the pathogenesis of lung cancer: a cell model for chronic exposure to cigarette smoke extract. BMC Pulm Med 2022; 22:333. [PMID: 36056339 PMCID: PMC9438261 DOI: 10.1186/s12890-022-02116-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Background Human tumors are highly heterogeneous at the cellular, molecular, genetic and functional levels. Tumor heterogeneity has tremendous impact on cancer progression and treatment responses. However, the mechanisms for tumor heterogeneity have been poorly understood due to the lack of experimental models. Methods This study provides a novel exploration and analysis of the impacts of cellular and molecular heterogeneity of human lung epithelial cells on their malignant transformation following chronic exposure to cigarette smoke extracts. Results The ability of cigarette smoke extract (CSE) to cause malignant transformation of the human bronchial epithelial cells (16HBE) is dependent on the sizes of the cells. Epithelial-mesenchymal transition (EMT) plays an important role in this process. Mechanistically, CSE-induced malignant transformation of 16HBE cells was closely linked to the reduced relative telomere length of the larger 16HBE cells, thereby up-regulation of the expression of stemness genes. Conclusions These findings provide novel insights for understanding the impact of cellular heterogeneity in lung cancer development. The in vitro transformation model described in this study could be extrapolated to studying the pathogenesis of other malignancies, as well as for mechanistic studies that are not feasible in vivo. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-02116-6.
Collapse
Affiliation(s)
- Dong Xia
- Department of Toxicology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Jieyi Liu
- Department of Toxicology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Juanjuan Yong
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Xiang Li
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, 450001, Henan, People's Republic of China
| | - Weidong Ji
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Zhiqiang Zhao
- Department of Toxicology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Xiaohui Wang
- Department of Toxicology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Chen Xiao
- Department of Toxicology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Sai Wu
- Department of Toxicology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Huaixiang Liu
- Department of Toxicology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Heping Zhao
- Department of Toxicology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Yun He
- Department of Toxicology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
19
|
Li C, Zhang Z, Peng E, Peng J. Role of an Exosomes-Related lncRNAs Signature in Tumor Immune Microenvironment of Gastric Cancer. Front Cell Dev Biol 2022; 10:873319. [PMID: 35465325 PMCID: PMC9019506 DOI: 10.3389/fcell.2022.873319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/21/2022] [Indexed: 12/25/2022] Open
Abstract
Background: Exosomes plays a crucial role in intercellular communication of gastric cancer (GC), while long non-coding RNAs (lncRNAs) contributes to the tumorigenesis and progression of GC. This study aims to explore the prognostic exosomes-related lncRNAs of GC patients. Methods: Data of 375 GC patients were obtained from the TCGA database. The entire cohort was randomly divided into a training cohort and a validation cohort in a 2:1 ratio. Exosomes-related lncRNAs were identified by the Pearson correlation analysis with reported exosomes-related genes. LASSO Cox regression was used to construct the signature. Results: A prognostic signature consisting of 11 exosomes-related lncRNAs was identified, and patients with lower risk scores had a better prognosis than those with higher risk scores. ROC curves and multivariate Cox regression analysis showed that the signature was an independent risk factor for prognosis in both the training (HR: 3.254, 95% CI: 2.310–4.583) and validation cohorts (HR: 1.974, 95% CI: 1.108–3.517). Gene set enrichment analysis (GSEA) suggested associations between the signature and several immune-related pathways. The identified signature was shown to be associated with GC tumor microenvironment. The expression of two immune checkpoints was also increased in the high-risk group, including B7-H3 and VSIR, indicating the potential role of the identified signature in GC immunotherapies. Conclusion: A novel exosomes-related lncRNA signature, which may be associated with tumor immune microenvironment and potentially serve as an indicator for immunotherapy, has been identified to precisely predict the prognosis of GC patients.
Collapse
Affiliation(s)
- Chan Li
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Zhang
- Department of Thyroid Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Emin Peng
- Xiangya International Medical Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Emin Peng, ; Jinwu Peng,
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Xiangya Changde Hospital, Changde, China
- *Correspondence: Emin Peng, ; Jinwu Peng,
| |
Collapse
|
20
|
Faghfuri E, Shadbad MA, Faghfouri AH, Soozangar N. Cellular immunotherapy in gastric cancer: adoptive cell therapy and dendritic cell-based vaccination. Immunotherapy 2022; 14:475-488. [PMID: 35232264 DOI: 10.2217/imt-2021-0285] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of the most frequently diagnosed malignancies. Recent studies have highlighted cellular immunotherapy (CI) as a promising approach for treating this disease. Among the CI-based approaches, adoptive cell therapy and dendritic cell-based vaccination are commonly studied in preclinical and clinical trials. Here we review the current evidence on the potentiality of CI in treating GC, the targets for adoptive cell therapy, ongoing clinical trials, constraints and the future outlook. The results suggest that there is a need to identify novel biomarkers that predict which GC patients will most likely respond to these approaches. Also, CI plus chemotherapy or immune checkpoint inhibitors can improve the survival of patients with late-stage GC. Therefore, this approach can be promising for treating these patients.
Collapse
Affiliation(s)
- Elnaz Faghfuri
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | | | - Narges Soozangar
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
21
|
Yu Z, Lin S, Xia F, Liu Y, Zhang D, Wang F, Wang Y, Li Q, Niu J, Cao C, Cui D, Sheng N, Ren J, Wang Z, Chen D. ExoSD chips for high-purity immunomagnetic separation and high-sensitivity detection of gastric cancer cell-derived exosomes. Biosens Bioelectron 2021; 194:113594. [PMID: 34474280 DOI: 10.1016/j.bios.2021.113594] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/29/2021] [Accepted: 08/25/2021] [Indexed: 12/23/2022]
Abstract
Gastric cancer cell-derived exosomes as biomarkers have a very high application potential to the non-invasive detection of early-stage gastric cancer. However, the small size of exosomes (30-150 nm) results in huge challenges in separating and detecting them from complex media (e.g., plasma, urine, saliva, and cell culture supernatant). Here we proposed a highly integrated exosome separation and detection (ExoSD) chip to immunomagnetic separate exosomes from cell culture supernatant in a manner of continuous flow, and to immunofluorescence detect gastric cancer cell-derived exosomes with high sensitivity. The ExoSD chip has achieved a high exosome recovery (>80%) and purity (>83%) at the injection rate of 4.8 mL/h. Furthermore, experimental results based on clinical serum samples of patients with gastric cancer (stages I and II) show that the detection rate of the ExoSD chip is as high as 70%. The proposed ExoSD chip has been successfully demonstrated as a cutting-edge platform for exosomes separation and detection. It can be served as a versatile platform to extend to the applications of separation and detection of the other cell-derived exosomes or cells.
Collapse
Affiliation(s)
- Zixian Yu
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai, 200240, PR China; Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, Shanghai, 200240, PR China
| | - Shujing Lin
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai, 200240, PR China; Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, Shanghai, 200240, PR China.
| | - Fangfang Xia
- The Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicin, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Yanlei Liu
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai, 200240, PR China; Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, Shanghai, 200240, PR China
| | - Di Zhang
- Center for Advanced Electronic Materials and Devices (AEMD), Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Fei Wang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai, 200240, PR China; Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, Shanghai, 200240, PR China
| | - Yanpu Wang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai, 200240, PR China; Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, Shanghai, 200240, PR China
| | - Qichao Li
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai, 200240, PR China; Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, Shanghai, 200240, PR China
| | - Jiaqi Niu
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai, 200240, PR China; Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, Shanghai, 200240, PR China
| | - Chengxi Cao
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai, 200240, PR China
| | - Daxiang Cui
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai, 200240, PR China; Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, Shanghai, 200240, PR China
| | - Nengquan Sheng
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China
| | - Jiazi Ren
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China
| | - Zhigang Wang
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China.
| | - Di Chen
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai, 200240, PR China; Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, Shanghai, 200240, PR China.
| |
Collapse
|
22
|
Ben Rejeb S, Beltaifa D, Ghozzi A, Bellil K, Turki S. EpCAM (MOC-31) - immunohistochemical expression in papillary thyroid carcinoma and non invasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP). LA TUNISIE MEDICALE 2021; 99:1066-1071. [PMID: 35288910 PMCID: PMC8974436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Ep-CAM, is a cell adhesion glycoprotein located on the basolateral cell membrane surface and in the cytoplasm of most normal epithelial cells. It has also been described to be expressed in several malignancies such as lung, digestive, prostate and renal carcinomas suggesting it has a potential role in carcinogenesis. In thyroid carcinoma, Ep-CAM expression has rarely been studied especially in papillary thyroid carcinoma. OBJECTIVE We sought to describe and compare the immunohistochemical expression of MOC31 in papillary thyroid carcinoma and in non invasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP). METHODS We have retrospectively collected 33 cases of PTC diagnosed in the pathology department of the Security forces hospital during a period of 13 years (2008-2021). We have microscopically reviewed all cases and reclassified 9 of 33 cases as NIFTP. An immunohistochemical automated study have been performed with MOC-31 antibody. The immunostaining was considered positive when it was membranous and/or cytoplasmic. The intensity of staining was scored as weak (score 1), moderate (score 2), and strong (score 3). We have used an immunoscore for assessing level of expression of MOC31 as follows: 0 for <5% of positive cells, 1 for 5-30%, 2 for 31-50%, 3 for 51-70%.The total score resulted by summing the percentage score with the intensity score; the final score was varying from 0 to 7, considered low between 1-4 and high 5-7. RESULTS The mean age of patients was 45,2 years-old for PTC cases and 48,1 years-old for NIFTP cases. A net female predominance was found in both groups (male to female ratio of respectively 0,4 and 0,3). MOC31 expression was found in 19 cases of PTC with a percentage of positive cells varying from 5 to 90%. Percentage of positive cells was variable from 5 to 90%. The immunoscore for positive cells was: 0 in 5/24cases, 1 in 4/24cases, 3 in 9/24cases and 4 in 6/24cases. The intensity of staining was assessed score2 (moderate) in 8 cases and score 3 (high) in 7cases (Figure1-2). Final MOC31 staining score was low in 37,5% (9/24) and high in 62.5% (15/24). Patients with advanced pt2-pt3 stages mostly showed high score of MOC31 staining (61,5%).One case was associated with lymph node involvement and was of a high score. 6 cases showed vascular invasion and was of high MOC31 score. MOC31 was expressed in all NIFTP cases with variable proportion of positive cells (5%-80%). The immunoscore for positive cells was: 0 in 1/9cases, 1 in 2/9cases, 2 in 3/9cases, 3 in 1/9cases and 4 in 2/9cases. The intensity of staining was assessed score 1 (weak) in one case, score 2 (moderate) in 6 cases and score 3 (high) in one case (Figure3-4). The final combined score was low in 66,7 (6/9) and high in 33,3% (3/9). CONCLUSION Our study revealed different immunohistochemical profile of MOC31 in benign and malignant tumors. It has somewhat a diffuse and marked staining in the first group. The changes of MOC31 location as well as its score of staining in PTC and NIFTP could hence be helpful in the differential diagnosis. Our findings also support the potential prognostic value of this molecule that deserves further investigations.
Collapse
Affiliation(s)
- Sarra Ben Rejeb
- Hôpital des Forces de Sécurité Intérieure, Faculté de médecine de Tunis
| | - Dorsaf Beltaifa
- Hôpital des Forces de Sécurité Intérieure, Faculté de médecine de Tunis
| | - Amen Ghozzi
- Hôpital des Forces de Sécurité Intérieure, Faculté de médecine de Tunis
| | - Khadija Bellil
- Hôpital des Forces de Sécurité Intérieure, Faculté de médecine de Tunis
| | - Senda Turki
- Hôpital des Forces de Sécurité Intérieure, Faculté de médecine de Tunis
| |
Collapse
|
23
|
Olnes MJ, Martinson HA. Recent advances in immune therapies for gastric cancer. Cancer Gene Ther 2021; 28:924-934. [PMID: 33664460 PMCID: PMC8417143 DOI: 10.1038/s41417-021-00310-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/28/2021] [Accepted: 02/11/2021] [Indexed: 01/31/2023]
Abstract
Gastric cancer (GC) is an aggressive malignancy that is the third leading cause of cancer mortality worldwide. Localized GC can be cured with surgery, but most patients present with more advanced non-operable disease. Until recently, treatment options for relapsed and refractory advanced GC have been limited to combination chemotherapy regimens, HER-2 directed therapy, and radiation, which lead to few durable responses. Over the past decade, there have been significant advances in our understanding of the molecular and immune pathogenesis of GC. The infectious agents Epstein-Barr virus and Helicobacter pylori perturb the gastric mucosa immune equilibrium, which creates a microenvironment that favors GC tumorigenesis and evasion of immune surveillance. Insights into immune mechanisms of GC have translated into novel therapeutics, including immune checkpoint inhibitors, which have become a treatment option for select patients with GC. Furthermore, chimeric antigen receptor T-cell therapies have emerged as a breakthrough treatment for many cancers, with recent studies showing this to be a potential therapy for GC. In this review, we summarize the current state of knowledge on immune mechanisms of GC and the status of emerging immunotherapies to treat this aggressive cancer, as well as outline current challenges and directions for future research.
Collapse
Affiliation(s)
- Matthew J Olnes
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, Anchorage, AK, USA.
- WWAMI School of Medical Education, University of Alaska Anchorage, Anchorage, AK, USA.
| | - Holly A Martinson
- WWAMI School of Medical Education, University of Alaska Anchorage, Anchorage, AK, USA
| |
Collapse
|
24
|
Toh J, Hoppe MM, Thakur T, Yang H, Tan KT, Pang B, Ho S, Roy R, Ho KY, Yeoh KG, Tan P, Sundar R, Jeyasekharan A. Profiling of gastric cancer cell-surface markers to achieve tumour-normal discrimination. BMJ Open Gastroenterol 2021; 7:bmjgast-2020-000452. [PMID: 32816956 PMCID: PMC7437876 DOI: 10.1136/bmjgast-2020-000452] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/04/2020] [Accepted: 07/17/2020] [Indexed: 12/24/2022] Open
Abstract
Background Differentiating between malignant and normal cells within tissue samples is vital for molecular profiling of cancer using advances in genomics and transcriptomics. Cell-surface markers of tumour–normal discrimination have additional value in terms of translatability to diagnostic and therapeutic strategies. In gastric cancer (GC), previous studies have identified individual genes or proteins that are upregulated in cancer. However, a systematic analysis of cell-surface markers and development of a composite panel involving multiple candidates to differentiate tumour from normal has not been previously reported. Methods Whole transcriptome sequencing (WTS) of GC and matched normal samples from the Singapore Gastric Cancer Consortium (SGCC) was used as a discovery cohort to identify upregulated putative cell-surface proteins. Matched WTS data from the The Cancer Genome Atlas (TCGA) was used as a validation cohort. Promising candidates from this analysis were validated orthogonally using multispectral immunohistochemistry (mIHC) with automated quantitative analysis using the Vectra platform. mIHC was performed on a tissue microarray containing matched normal, marginal and tumour tissues. The receiver-operating characteristic (ROC) curves were analysed to identify markers with the highest diagnostic validity independently and in combination. Results Analysis of putative membrane protein transcripts from the SGCC discovery cohort WTS data (n=15 matched tumour and normal pairs) identified several differentially and highly expressed candidates in tumour compared with normal tissues. After validation with TCGA data (n=29 matched tumour and normal pairs), the following proteins were selected for mIHC analysis: CEACAM5, CEACAM6, CLDN4, CLDN7, and EpCAM. These were compared with established glycoprotein markers in GC, namely CA19-9 and CA72-4. Individual ROC curves yielded the best performance for CEACAM5 (area under the ROC curve (AUC)=0.80), CEACAM6 (AUC=0.82), EpCAM (AUC=0.83), and CA72-4 (AUC=0.76). Combined multiplexed imaging of these four markers revealed improved specificity and sensitivity for detection of tumour from normal tissue (AUC of 4-plex=0.91). Conclusion CEAMCAM5, CEACAM6, EpCAM, and CA72-4 form a versatile set of markers for robust discrimination of GC from adjacent normal tissue. As cell-surface markers, they are compatible with both IHC and live imaging approaches. These candidates may be exploited to improve automated identification of tumour tissue in GC.
Collapse
Affiliation(s)
- Judith Toh
- Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Michal Marek Hoppe
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Teena Thakur
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Kar Tong Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Brendan Pang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Sharmaine Ho
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Rony Roy
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Khek Yu Ho
- Yong Loo Lin School of Medicine, National University Singapore, Singapore.,Medicine, Gastroenterology & Hepatology, National University Hospital, Singapore
| | - Khay Guan Yeoh
- Yong Loo Lin School of Medicine, National University Singapore, Singapore.,Department of Medicine, National University of Singapore and Senior Consultant Gastroenterologist, Singapore
| | - Patrick Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.,Agency for Science, Technology and Research, Genome Institute of Singapore, Singapore.,SingHealth/ Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore
| | - Raghav Sundar
- Yong Loo Lin School of Medicine, National University Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, National University Cancer Institute, Singapore.,The N.1 Institute for Health, National University of Singapore, Singapore
| | - Anand Jeyasekharan
- Yong Loo Lin School of Medicine, National University Singapore, Singapore .,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, National University Cancer Institute, Singapore
| |
Collapse
|
25
|
Lin T, Peng W, Mai P, Zhang E, Peng L. Human Gastric Cancer Stem Cell (GCSC) Markers Are Prognostic Factors Correlated With Immune Infiltration of Gastric Cancer. Front Mol Biosci 2021; 8:626966. [PMID: 34113647 PMCID: PMC8185345 DOI: 10.3389/fmolb.2021.626966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/08/2021] [Indexed: 12/24/2022] Open
Abstract
The prognosis of patients with gastric cancer (GC) is still unsatisfying. Numerous markers of gastric cancer stem cells (GCSCs) have been identified and were thought to be related to cancer aggressiveness. However, the roles of GCSC markers in GC patients’ prognosis and immune infiltration remain unknown. Expression of GCSC markers was analyzed using Oncomine and Gene Expression Profiling Interactive Analysis (GEPIA). Their associations with clinicopathological parameters were analyzed using UALCAN and LinkedOmics. Alternations and protein expression of GCSC markers were analyzed by cBioPortal and the Human Protein Atlas databases, respectively. The prognostic significance of GCSC markers was evaluated using Kaplan-Meier plotter. Correlations between the expression of GCSC markers and immune infiltration along with biomarkers of tumor-infiltrating immune cells (TIICs) were assessed combined Tumor Immune Estimation Resource and GEPIA. GeneMANIA was used to discover the interactive genes of GCSC markers, and enrichment analysis was performed using Database for Annotation, Visualization, and Integrated Discovery server. We identified six GCSC markers significantly up-expressed in GC, compared with normal stomach tissues. Among them, the overexpression of ICAM1, THY1, and CXCR4 significantly indicated adverse, while EPCAM indicated beneficial clinicopathological features of GC patients. The up-regulation of CXCR4 showed unfavorable prognostic significance, whereas EPCAM and TFRC showed the opposite. The six GCSC markers were all correlated with the infiltration and activation of distinct TIICs. Especially, ICAM1, THY1, and CXCR4 showed strongly positive correlations with tumor-associated macrophages. Besides, chemokine, Toll-like receptor, NF-kappa B, and HIF-1 signaling pathways might be involved in the regulation of GCSC markers on cancer development. This study proposed that GCSC markers might be promising targets of GC treatment to weaken cancer stem-like properties and strengthen anticancer immunity.
Collapse
Affiliation(s)
- Tong Lin
- The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Wenya Peng
- The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Peipei Mai
- The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - E Zhang
- The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lisheng Peng
- Department of Science and Education, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
26
|
Wang C, Chen S, Wu Y, Wu D, Wang J, Li F. The combination therapy with EpCAM/CD3 BsAb and MUC-1/CD3 BsAb elicited antitumor immunity by T-cell adoptive immunotherapy in lung cancer. Int J Med Sci 2021; 18:3380-3388. [PMID: 34522164 PMCID: PMC8436090 DOI: 10.7150/ijms.61681] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/22/2021] [Indexed: 11/07/2022] Open
Abstract
Lung cancer remains a global challenge due to high morbidity and mortality rates and poor response to treatment, and there are still no effective strategies to solve it. The bispecific antibody (BsAb) is a novel antibody, which can target two different antigens and mediate specific killing effects by selectively redirecting effector cells to the target cells. In this study, we combined two BsAbs to achieve a dual-target therapy strategy of EpCAM+ and MUC-1+ with high affinity and specificity. The results showed that the combination of two BsAbs against EpCAM and MUC-1 could inhibit the growth of lung cancer more effectively in cell lines and primary tumors. The superior antitumor effect of two BsAbs could be attributable to enhanced CTL and increased production of type I IFNs. At the same time, the combination of EpCAM/CD3 BsAb and MUC-1/CD3 BsAb significantly regulated T population in the TDLNs. Therefore, we have found a potential immunotherapeutic strategy, which was the combination therapy with EpCAM/CD3 BsAb and MUC-1/CD3 BsAb for the treatment of non-small cell lung cancer.
Collapse
Affiliation(s)
- Ce Wang
- Shenzhen key laboratory of stem cell research and clinical transformation, Guangdong Engineering Technology Research Center of Stem cell and Cell therapy, Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Shang Chen
- Shenzhen key laboratory of stem cell research and clinical transformation, Guangdong Engineering Technology Research Center of Stem cell and Cell therapy, Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Yingjuan Wu
- Shenzhen key laboratory of stem cell research and clinical transformation, Guangdong Engineering Technology Research Center of Stem cell and Cell therapy, Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Di Wu
- Shenzhen key laboratory of stem cell research and clinical transformation, Guangdong Engineering Technology Research Center of Stem cell and Cell therapy, Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Jingbo Wang
- Shenzhen key laboratory of stem cell research and clinical transformation, Guangdong Engineering Technology Research Center of Stem cell and Cell therapy, Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Furong Li
- Shenzhen key laboratory of stem cell research and clinical transformation, Guangdong Engineering Technology Research Center of Stem cell and Cell therapy, Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| |
Collapse
|
27
|
Ghasemi F, Tessier TM, Gameiro SF, Maciver AH, Cecchini MJ, Mymryk JS. High MHC-II expression in Epstein-Barr virus-associated gastric cancers suggests that tumor cells serve an important role in antigen presentation. Sci Rep 2020; 10:14786. [PMID: 32901107 PMCID: PMC7479113 DOI: 10.1038/s41598-020-71775-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023] Open
Abstract
EBV-associated gastric adenocarcinomas (EBVaGCs) often exhibit better clinical outcomes than EBV negative gastric cancers (GCs), which could be related to their consistent expression of foreign viral antigens. Antigen-presenting cells (APCs) present peptide antigens in the context of the class-II major histocompatibility complex (MHC-II). During inflammatory conditions, epithelial cells express MHC-II and function as accessory APCs. Utilizing RNA-seq data from nearly 400 GC patients, we determined the impact of EBV-status on expression of MHC-II components, genes involved in their regulation, and T-cell co-stimulation. Virtually all MHC-II genes were significantly upregulated in EBVaGCs compared to normal tissues, or other GC subtypes. Genes involved in antigen presentation were also significantly upregulated in EBVaGCs, as were the key MHC-II transcriptional regulators CIITA and RFX5. This was unexpected as the EBV encoded BZLF1 protein can repress CIITA transcription and is expressed in many EBVaGCs. Furthermore, MHC-II upregulation was strongly correlated with elevated intratumoral levels of interferon-gamma. In addition, expression of co-stimulatory molecules involved in T-cell activation and survival was also significantly increased in EBVaGCs. Thus, gastric adenocarcinoma cells may functionally contribute to the highly immunogenic tumor microenvironment observed in EBVaGCs via a previously unappreciated role in interferon-induced antigen presentation.
Collapse
Affiliation(s)
- Farhad Ghasemi
- Department of Surgery, Western University, London, ON, N6A 4V2, Canada
| | - Tanner M Tessier
- Department of Microbiology and Immunology, Western University, London, ON, N6A 3K7, Canada
| | - Steven F Gameiro
- Department of Microbiology and Immunology, Western University, London, ON, N6A 3K7, Canada
| | - Allison H Maciver
- Department of Surgery, Western University, London, ON, N6A 4V2, Canada.,Department of Oncology, Western University, London, ON, N6A 3K7, Canada
| | - Matthew J Cecchini
- Department of Pathology and Laboratory Medicine, Western University and London Health Sciences Centre, London, ON, N6A 5C1, Canada
| | - Joe S Mymryk
- Department of Microbiology and Immunology, Western University, London, ON, N6A 3K7, Canada. .,Department of Oncology, Western University, London, ON, N6A 3K7, Canada. .,Department of Otolaryngology, Head & Neck Surgery, Western University, London, ON, N6A 5W9, Canada. .,London Regional Cancer Program, Lawson Health Research Institute, London, ON, N6C 2R5, Canada. .,London Regional Cancer Program, Room A4-837, 790 Commissioners Rd. East, London, ON, N6A 4L6, Canada.
| |
Collapse
|
28
|
Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, Blaudszun AR, Yevsa T, Fricke S, Kossatz-Boehlert U. Cancer Stem Cells-Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Front Immunol 2020; 11:1280. [PMID: 32849491 PMCID: PMC7426526 DOI: 10.3389/fimmu.2020.01280] [Citation(s) in RCA: 541] [Impact Index Per Article: 108.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023] Open
Abstract
The use of biomarkers in diagnosis, therapy and prognosis has gained increasing interest over the last decades. In particular, the analysis of biomarkers in cancer patients within the pre- and post-therapeutic period is required to identify several types of cells, which carry a risk for a disease progression and subsequent post-therapeutic relapse. Cancer stem cells (CSCs) are a subpopulation of tumor cells that can drive tumor initiation and can cause relapses. At the time point of tumor initiation, CSCs originate from either differentiated cells or adult tissue resident stem cells. Due to their importance, several biomarkers that characterize CSCs have been identified and correlated to diagnosis, therapy and prognosis. However, CSCs have been shown to display a high plasticity, which changes their phenotypic and functional appearance. Such changes are induced by chemo- and radiotherapeutics as well as senescent tumor cells, which cause alterations in the tumor microenvironment. Induction of senescence causes tumor shrinkage by modulating an anti-tumorigenic environment in which tumor cells undergo growth arrest and immune cells are attracted. Besides these positive effects after therapy, senescence can also have negative effects displayed post-therapeutically. These unfavorable effects can directly promote cancer stemness by increasing CSC plasticity phenotypes, by activating stemness pathways in non-CSCs, as well as by promoting senescence escape and subsequent activation of stemness pathways. At the end, all these effects can lead to tumor relapse and metastasis. This review provides an overview of the most frequently used CSC markers and their implementation as biomarkers by focussing on deadliest solid (lung, stomach, liver, breast and colorectal cancers) and hematological (acute myeloid leukemia, chronic myeloid leukemia) cancers. Furthermore, it gives examples on how the CSC markers might be influenced by therapeutics, such as chemo- and radiotherapy, and the tumor microenvironment. It points out, that it is crucial to identify and monitor residual CSCs, senescent tumor cells, and the pro-tumorigenic senescence-associated secretory phenotype in a therapy follow-up using specific biomarkers. As a future perspective, a targeted immune-mediated strategy using chimeric antigen receptor based approaches for the removal of remaining chemotherapy-resistant cells as well as CSCs in a personalized therapeutic approach are discussed.
Collapse
Affiliation(s)
- Lia Walcher
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Ann-Kathrin Kistenmacher
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Huizhen Suo
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Reni Kitte
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Sarah Dluczek
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Alexander Strauß
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - André-René Blaudszun
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Tetyana Yevsa
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Stephan Fricke
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Uta Kossatz-Boehlert
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
29
|
CAR-T Cell Therapy-An Overview of Targets in Gastric Cancer. J Clin Med 2020; 9:jcm9061894. [PMID: 32560392 PMCID: PMC7355670 DOI: 10.3390/jcm9061894] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is one of the most commonly diagnosed malignancies and, unfortunately, still has a high mortality rate. Recent research points to CAR-T immunotherapy as a promising treatment for this disease. Using genetically engineered T cells designed to target a previously selected antigen, researchers are able to harness the natural anti-tumor activity of T cells. For therapy to be successful, however, it is essential to choose antigens that are present on tumor cells but not on healthy cells. In this review, we present an overview of the most important targets for CAR-T therapy in the context of GC, including their biologic function and therapeutic application. A number of clinical studies point to the following as important markers in GC: human epidermal growth factor receptor 2, carcinoembryonic antigen, mucin 1, epithelial cell adhesion molecule, claudin 18.2, mesothelin, natural-killer receptor group 2 member D, and folate receptor 1. Although these markers have been met with some success, the search for new and improved targets continues. Key among these novel biomarkers are the B7H6 ligand, actin-related protein 2/3 (ARP 2/3), neuropilin-1 (NRP-1), desmocollin 2 (DSC2), anion exchanger 1 (AF1), and cancer-related antigens CA-72-4 and CA-19-9.
Collapse
|
30
|
Matrix Effect in the Isolation of Breast Cancer-Derived Nanovesicles by Immunomagnetic Separation and Electrochemical Immunosensing-A Comparative Study. SENSORS 2020; 20:s20040965. [PMID: 32054015 PMCID: PMC7071381 DOI: 10.3390/s20040965] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 01/16/2023]
Abstract
Exosomes are cell-derived nanovesicles released into biological fluids, which are involved in cell-to-cell communication. The analysis of the content and the surface of the exosomes allow conclusions about the cells they are originating from and the underlying condition, pathology or disease. Therefore, the exosomes are currently considered good candidates as biomarkers to improve the current methods for clinical diagnosis, including cancer. However, due to their low concentration, conventional procedures for exosome detection including biosensing usually require relatively large sample volumes and involve preliminary purification and preconcentration steps by ultracentrifugation. In this paper, the immunomagnetic separation is presented as an alternative method for the specific isolation of exosomes in serum. To achieve that, a rational study of the surface proteins in exosomes, which can be recognized by magnetic particles, is presented. The characterization was performed in exosomes obtained from cell culture supernatants of MCF7, MDA-MB-231 and SKBR3 breast cancer cell lines, including TEM and nanoparticle tracking analysis (NTA). For the specific characterization by flow cytometry and confocal microscopy, different commercial antibodies against selected receptors were used, including the general tetraspanins CD9, CD63 and CD81, and cancer-related receptors (CD24, CD44, CD54, CD326 and CD340). The effect of the serum matrix on the immunomagnetic separation was then carefully evaluated by spiking the exosomes in depleted human serum. Based on this study, the exosomes were preconcentrated by immunomagnetic separation on antiCD81-modified magnetic particles in order to achieve further magnetic actuation on the surface of the electrode for the electrochemical readout. The performance of this approach is discussed and compared with classical characterization methods.
Collapse
|
31
|
Si M, Zhang J, Cao J, Xie Z, Shu S, Zhu Y, Lang J. Integrated Analysis To Identify Molecular Biomarkers Of High-Grade Serous Ovarian Cancer. Onco Targets Ther 2019; 12:10057-10075. [PMID: 31819501 PMCID: PMC6877452 DOI: 10.2147/ott.s228678] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Abstract
Purpose Ovarian cancer is the leading cause of gynecologic cancer-related death worldwide. Early diagnosis of ovarian cancer can significantly improve patient prognosis. Hence, there is an urgent need to identify key diagnostic and prognostic biomarkers specific for ovarian cancer. Because high-grade serous ovarian cancer (HGSOC) is the most common type of ovarian cancer and accounts for the majority of deaths, we identified potential biomarkers for the early diagnosis and prognosis of HGSOC. Methods Six datasets (GSE14001, GSE18520, GSE26712, GSE27651, GSE40595, and GSE54388) were downloaded from the Gene Expression Omnibus database for analysis. Differentially expressed genes (DEGs) between HGSOC and normal ovarian surface epithelium samples were screened via integrated analysis. Hub genes were identified by analyzing protein-protein interaction (PPI) network data. The online Kaplan-Meier plotter was utilized to evaluate the prognostic roles of these hub genes. The expression of these hub genes was confirmed with Oncomine datasets and validated by quantitative real-time PCR and Western blotting. Results A total of 103 DEGs in patients with HGSOC-28 upregulated genes and 75 downregulated genes-were successfully screened. Enrichment analyses revealed that the upregulated genes were enriched in cell division and cell proliferation and that the downregulated genes mainly participated in the Wnt signaling pathway and various metabolic processes. Ten hub genes were associated with HGSOC pathogenesis. Seven overexpressed hub genes were partitioned into module 1 of the PPI network, which was enriched in the cell cycle and DNA replication pathways. Survival analysis revealed that MELK, CEP55 and KDR expression levels were significantly correlated with the overall survival of HGSOC patients (P < 0.05). The RNA and protein expression levels of these hub genes were validated experimentally. Conclusion Based on an integrated analysis, we propose the further investigation of MELK, CEP55 and KDR as promising diagnostic and prognostic biomarkers of HGSOC.
Collapse
Affiliation(s)
- Manfei Si
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Junji Zhang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Jianzhong Cao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Zhibo Xie
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Shan Shu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yapei Zhu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
32
|
Tan HL, Choo A. Opportunities for Antibody Discovery Using Human Pluripotent Stem Cells: Conservation of Oncofetal Targets. Int J Mol Sci 2019; 20:E5752. [PMID: 31731794 PMCID: PMC6888136 DOI: 10.3390/ijms20225752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Pluripotent stem cells (PSCs) comprise both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). The application of pluripotent stem cells is divided into four main areas, namely: (i) regenerative therapy, (ii) the study and understanding of developmental biology, (iii) drug screening and toxicology and (iv) disease modeling. In this review, we describe a new opportunity for PSCs, the discovery of new biomarkers and generating antibodies against these biomarkers. PSCs are good sources of immunogen for raising monoclonal antibodies (mAbs) because of the conservation of oncofetal antigens between PSCs and cancer cells. Hence mAbs generated using PSCs can potentially be applied in two different fields. First, these mAbs can be used in regenerative cell therapy to characterize the PSCs. In addition, the mAbs can be used to separate or eliminate contaminating or residual undifferentiated PSCs from the differentiated cell product. This step is critical as undifferentiated PSCs can form teratomas in vivo. The mAbs generated against PSCs can also be used in the field of oncology. Here, novel targets can be identified and the mAbs developed as targeted therapy to kill the cancer cells. Conversely, as new and novel oncofetal biomarkers are discovered on PSCs, cancer mAbs that are already approved by the FDA can be repurposed for regenerative medicine, thus expediting the route to the clinics.
Collapse
Affiliation(s)
- Heng Liang Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore 138668, Singapore;
| | - Andre Choo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore 138668, Singapore;
- Department of Biochemical Engineering, National University of Singapore, Singapore 117575, Singapore
| |
Collapse
|
33
|
Pape J, Magdeldin T, Ali M, Walsh C, Lythgoe M, Emberton M, Cheema U. Cancer invasion regulates vascular complexity in a three-dimensional biomimetic model. Eur J Cancer 2019; 119:179-193. [PMID: 31470251 DOI: 10.1016/j.ejca.2019.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/03/2019] [Accepted: 07/05/2019] [Indexed: 11/20/2022]
Abstract
INTRODUCTION There is a growing appreciation for including a complex, vascularised stroma in three-dimensional (3D) tumour models to recapitulate the native tumour microenvironment in situ. METHODS Using a compartmentalised, biomimetic, 3D cancer model, comprising a central cancer mass surrounded by a vascularised stroma, we have tested the invasive capability of colorectal cancer cells. RESULTS We show histological analysis of dense collagen I/laminin scaffolds, forming necrotic cores with cellular debris. Furthermore, cancer cells within this 3D matrix form spheroids, which is corroborated with high EpCAM expression. We validate the invasive growth of cancer cells into the stroma through quantitative image analysis and upregulation of known invasive gene markers, including metastasis associated in colon cancer 1, matrix metalloproteinase 7 and heparinase. Tumouroids containing highly invasive HCT116 cancer masses form less complex and less branched vascular networks, recapitulating 'leaky' vasculature associated with highly metastatic cancers. Angiogenic factors regulating this were vascular endothelial growth factor A and hepatocyte growth factor active protein. Where vascular networks were formed with less invasive cancer masses (HT29), higher expression of vascular endothelial cadherin active protein resulted in more complex and branched networks. To eliminate the cell-cell interaction between the cancer mass and stroma, we developed a three-compartment model containing an acellular ring to test the chemoattractant pull from the cancer mass. This resulted in migration of endothelial networks through the acellular ring accompanied by alignment of vascular networks at the cancer/stroma boundary. DISCUSSION This work interrogates to the gene and protein level how cancer cells influence the development of a complex stroma, which shows to be directly influenced by the invasive capability of the cancer.
Collapse
Affiliation(s)
- Judith Pape
- Institute of Orthopaedics and Musculoskeletal Sciences, Division of Surgery and Interventional Science, University College London, Stanmore Campus, Brockley Hill, HA7 4LP, London, United Kingdom
| | - Tarig Magdeldin
- Institute of Orthopaedics and Musculoskeletal Sciences, Division of Surgery and Interventional Science, University College London, Stanmore Campus, Brockley Hill, HA7 4LP, London, United Kingdom
| | - Morium Ali
- Center for Advanced Biomedical Imaging, Paul O'Gorman Building, 72 Huntley Street, University College London, WC1E 6DD, London, United Kingdom
| | - Claire Walsh
- Center for Advanced Biomedical Imaging, Paul O'Gorman Building, 72 Huntley Street, University College London, WC1E 6DD, London, United Kingdom
| | - Mark Lythgoe
- Center for Advanced Biomedical Imaging, Paul O'Gorman Building, 72 Huntley Street, University College London, WC1E 6DD, London, United Kingdom
| | - Mark Emberton
- Faculty of Medical Sciences, University College London, Bloomsbury Campus Maple House, 149 Tottenham Court Road, W1T 7NF, London, United Kingdom
| | - Umber Cheema
- Institute of Orthopaedics and Musculoskeletal Sciences, Division of Surgery and Interventional Science, University College London, Stanmore Campus, Brockley Hill, HA7 4LP, London, United Kingdom.
| |
Collapse
|
34
|
Hu Y, Wu Q, Gao J, Zhang Y, Wang Y. A meta-analysis and The Cancer Genome Atlas data of prostate cancer risk and prognosis using epithelial cell adhesion molecule (EpCAM) expression. BMC Urol 2019; 19:67. [PMID: 31324239 PMCID: PMC6642570 DOI: 10.1186/s12894-019-0499-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/09/2019] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Epithelial cell adhesion molecule (EpCAM) expression has been reported in many types of cancer, including prostate cancer (PCa). However, the role of EpCAM expression remains inconsistent. We conducted a meta-analysis to assess the clinicopathological and prognostic significance of EpCAM expression in PCa. METHODS Publications were searched online using electronic databases. The available data were obtained from The Cancer Genome Atlas (TCGA). The odds ratios (ORs) or hazard ratios (HRs) with their 95% confidence intervals (CIs) were calculated. RESULTS We identified seven studies in which immunohistochemistry was used and that included 871 prostatic tissue samples. EpCAM expression was significantly higher in PCa samples than in benign and normal tissue samples (OR = 77.93, P = 0.002; OR = 161.61, P < 0.001; respectively). No correlation of EpCAM overexpression with pT stage and lymph node metastasis was observed; however, EpCAM overexpression showed a significant correlation with Gleason score (OR = 0.48, P = 0.012) and bone metastasis (OR = 145.80, P < 0.001). Furthermore, TCGA data showed that EpCAM overexpression was not closely correlated with age, pT stage, lymph node metastasis, number of lymph node, prostate-specific antigen level, Gleason score, biochemical recurrence, and overall survival. Based on multivariate Cox proportional-hazards regression analysis, a significant correlation was observed between EpCAM overexpression and 5-year worse biochemical recurrence free-survival. CONCLUSIONS EpCAM overexpression may be correlated with the development of bone metastasis and worse biochemical recurrence free-survival of PCa. Further studies are needed to verify these findings.
Collapse
Affiliation(s)
- Yu Hu
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Qiong Wu
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Jialin Gao
- Department of Urology, The First Hospital of Jilin University, No. 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Yongrui Zhang
- Department of Urology, The First Hospital of Jilin University, No. 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Yuantao Wang
- Department of Urology, The First Hospital of Jilin University, No. 71 Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
35
|
Zhou Y, Wen P, Li M, Li Y, Li X. Construction of chimeric antigen receptor‑modified T cells targeting EpCAM and assessment of their anti‑tumor effect on cancer cells. Mol Med Rep 2019; 20:2355-2364. [PMID: 31322180 DOI: 10.3892/mmr.2019.10460] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 05/09/2019] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yan Zhou
- Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Ping Wen
- Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Mingmei Li
- Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Yaqi Li
- Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Xiao‑An Li
- Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
36
|
Xu P, Kai C, Kawasaki S, Kobayashi Y, Yamamoto K, Tsujikawa M, Hayashi R, Nishida K. A New in Vitro Model of GDLD by Knocking Out TACSTD2 and Its Paralogous Gene EpCAM in Human Corneal Epithelial Cells. Transl Vis Sci Technol 2018; 7:30. [PMID: 30619650 PMCID: PMC6314060 DOI: 10.1167/tvst.7.6.30] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/24/2018] [Indexed: 12/11/2022] Open
Abstract
Purpose Gelatinous drop-like corneal dystrophy (GDLD) is a rare autosomal recessive corneal dystrophy that causes severe vision loss. Because of its poor prognosis, there is a demand for novel treatments for GDLD. Here, we establish a new in vitro disease model of GDLD based on immortalized human corneal epithelial (HCE-T) cells. Methods By using transcription activator-like effector nuclease plasmids, tumor-associated calcium signal transducer 2 (TACSTD2) and its paralogous gene, epithelial cell adhesion molecule (EpCAM), were knocked out in HCE-T cells. Fluorescence-activated cell sorting was performed to obtain cells in which both TACSTD2 and EpCAM were knocked out (DKO cells). In DKO cells, the expression levels and subcellular localizations of claudin (CLDN) 1, 4, and 7, and ZO-1 were investigated, along with epithelial barrier function. By using DKO cells, the feasibility of gene therapy for GDLD was also investigated. Results DKO cells exhibited decreased expression and aberrant subcellular localization of CLDN1 and CLDN7 proteins, as well as decreased epithelial barrier function. Transduction of the TACSTD2 gene into DKO cells nearly normalized expression levels and subcellular localization of CLDN1 and CLDN7 proteins, while significantly increasing epithelial barrier function. Conclusions We established an in vitro disease model of GDLD by knocking out TACSTD2 and its paralogous gene, EpCAM, in HCE-T cells. This cell line accurately reflected pathological aspects of GDLD. Translational Relevance We expect that the cell line will be useful to elucidate the pathogenesis of GDLD and develop novel treatments for GDLD.
Collapse
Affiliation(s)
- Peng Xu
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Ocular Immunology and Regenerative Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chifune Kai
- Department of Ocular Immunology and Regenerative Medicine, Osaka University Graduate School of Medicine, Osaka, Japan.,Faculty of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Kawasaki
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Ocular Immunology and Regenerative Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuki Kobayashi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kouji Yamamoto
- Department of Biostatistics, Yokohama City University, School of Medicine, Kanagawa, Japan
| | - Motokazu Tsujikawa
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan.,Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryuhei Hayashi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
37
|
Tan HL, Yong C, Tan BZ, Fong WJ, Padmanabhan J, Chin A, Ding V, Lau A, Zheng L, Bi X, Yang Y, Choo A. Conservation of oncofetal antigens on human embryonic stem cells enables discovery of monoclonal antibodies against cancer. Sci Rep 2018; 8:11608. [PMID: 30072783 PMCID: PMC6072701 DOI: 10.1038/s41598-018-30070-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 06/26/2018] [Indexed: 12/26/2022] Open
Abstract
Monoclonal antibodies (mAbs) are used as targeted therapies against cancers. These mAbs kill cancer cells via various mechanisms of actions. In this study, human embryonic stem cells (hESCs) was used as the immunogen to generate a panel of antibodies. From this panel of mAbs, A19 was found to bind both hESC and various cancer cell lines. The antigen target of A19 was identified as Erbb-2 and glycan analysis showed that A19 binds to a N-glycan epitope on the antigen. A19 was elucidated to internalize into cancer cells following binding to Erbb-2 and hence developed as an antibody-drug conjugate (ADC). Using ADC as the mechanism of action, A19 was able to kill cancer cells in vitro and delayed the onset of tumour formation in mice xenograft model. When compared to Herceptin, A19 binds to different isoforms of Erbb-2 and does not compete with Herceptin for the same epitope. Hence, A19 has the potential to be developed as an alternative targeted therapeutic agent for cancers expressing Erbb-2.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Murine-Derived/immunology
- Antibodies, Monoclonal, Murine-Derived/pharmacology
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/immunology
- Antineoplastic Agents, Immunological/pharmacology
- Cell Line, Tumor
- Female
- Human Embryonic Stem Cells/immunology
- Humans
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Heng Liang Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore.
| | - Charlene Yong
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Bao Zhu Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Wey Jia Fong
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Jayanthi Padmanabhan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Angela Chin
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Vanessa Ding
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Ally Lau
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Lu Zheng
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Xuezhi Bi
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Yuansheng Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Andre Choo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| |
Collapse
|
38
|
Zhou L, Zhu Y. The EpCAM overexpression is associated with clinicopathological significance and prognosis in hepatocellular carcinoma patients: A systematic review and meta-analysis. Int J Surg 2018; 56:274-280. [DOI: 10.1016/j.ijsu.2018.06.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/07/2018] [Accepted: 06/10/2018] [Indexed: 02/08/2023]
|
39
|
Herreros-Pomares A, Aguilar-Gallardo C, Calabuig-Fariñas S, Sirera R, Jantus-Lewintre E, Camps C. EpCAM duality becomes this molecule in a new Dr. Jekyll and Mr. Hyde tale. Crit Rev Oncol Hematol 2018; 126:52-63. [DOI: 10.1016/j.critrevonc.2018.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 02/08/2023] Open
|
40
|
Boesch M, Spizzo G, Seeber A. Concise Review: Aggressive Colorectal Cancer: Role of Epithelial Cell Adhesion Molecule in Cancer Stem Cells and Epithelial-to-Mesenchymal Transition. Stem Cells Transl Med 2018; 7:495-501. [PMID: 29667344 PMCID: PMC5980125 DOI: 10.1002/sctm.17-0289] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/31/2018] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies worldwide. In spite of various attempts to ameliorate outcome by escalating treatment, significant improvement is lacking particularly in the adjuvant setting. It has been proposed that cancer stem cells (CSCs) and the epithelial‐to‐mesenchymal transition (EMT) are at least partially responsible for therapy resistance in CRC. The epithelial cell adhesion molecule (EpCAM) was one of the first CSC antigens to be described. Furthermore, an EpCAM‐specific antibody (edrecolomab) has the merit of having launched the era of monoclonal antibody treatment in oncology in the 1990s. However, despite great initial enthusiasm, monoclonal antibody treatment has not proven successful in the adjuvant treatment of CRC patients. In the meantime, new insights into the function of EpCAM in CRC have emerged and new drugs targeting various epitopes have been developed. In this review article, we provide an update on the role of EpCAM in CSCs and EMT, and emphasize the potential predictive selection criteria for novel treatment strategies and refined clinical trial design. stemcellstranslationalmedicine2018;7:495–501
Collapse
Affiliation(s)
- Maximilian Boesch
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute (TKFI), Innsbruck, Austria
| | - Gilbert Spizzo
- Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute (TKFI), Innsbruck, Austria
| | - Andreas Seeber
- Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute (TKFI), Innsbruck, Austria
| |
Collapse
|
41
|
Zhou YY, Kang YT, Chen C, Xu FF, Wang HN, Jin R. Combination of TNM staging and pathway based risk score models in patients with gastric cancer. J Cell Biochem 2018; 119:3608-3617. [PMID: 29231991 DOI: 10.1002/jcb.26563] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/28/2017] [Indexed: 01/18/2023]
Abstract
Due to the complexity and heterogeneity of gastric cancer (GC) in individual patient, current staging system is inadequate for predicting outcome of GC. Comprehensive computational and bioinformatics approach may triumph for the prediction. In this study, GC patients were devided according to stage and treatment: curative surgery plus chemoradiotherapy in stage II, curative surgery plus chemoradiotherapy in stages III, and IV, unresectable metastatic gastric cancer. The training sets were downloaded from GEO datasets (GSE26253 and GSE14208). Gene set enrichment analysis (GSEA) was performed to explore enriched difference between recurrence and nonrecurrence. The core enrichment genes of enriched pathways significantly associated with recurrence or progression were identified using Cox proportional hazards analysis. Thereafter, the risk score models were externally validated in independent datasets-GSE15081 and The Cancer Genome Atlas (TCGA). We generated respective risk score models of patients in different stages and treatment. A five-gene signature comprising FARP1, SGCE, SGCA, LAMA4, and COL9A2 was strongly associated with recurrence of patients with curative surgery plus chemoradiotherapy in stage II. A six-gene signature consisting of SHH, NF1, AP4B1, COMP, MATN3, and CCL8 was correlated with recurrence of patients with curative surgery plus chemoradiotherapy in stages III and IV. And a four-gene signature composing of ABCC2, AHNAK2, RNF43, and GSPT2 was highly related to progression of patients with unresectable metastatic GC. Taking into consideration TNM stage and gene signature reflecting recurrence or progression, the risk score models significantly improved the accuracy in predicting outcome of GC.
Collapse
Affiliation(s)
- Yang-Yang Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan-Ting Kang
- Department of Ultrasonography, Yichun people's hospital, Yichun, Jiangxi, China
| | - Chao Chen
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fan-Fan Xu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hao-Nan Wang
- School of Pharmaceutical sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rong Jin
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Epidemiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
42
|
Li K, Zhang Y, Chen M, Hu Y, Jiang W, Zhou L, Li S, Xu M, Zhao Q, Wan R. Enhanced antitumor efficacy of doxorubicin-encapsulated halloysite nanotubes. Int J Nanomedicine 2017; 13:19-30. [PMID: 29296083 PMCID: PMC5741065 DOI: 10.2147/ijn.s143928] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
To improve the antitumor efficacy of doxorubicin (DOX) and provide novel clinical treatment of gastric cancer, halloysite nanotubes (HNTs) loaded with DOX were encapsulated by soybean phospholipid (LIP) and the formed HNTs/DOX/LIP was systematically characterized via different techniques. The in vitro anticancer activity of HNTs/DOX/LIP was examined using an MTT assay. The antitumor efficacy and biocompatibility were monitored by measuring the tumor volume and assessing the blood routine and serum biochemistry using an ectopic implantation cancer model. The results show that when the concentration of HNTs was 3 mg/mL and the concentration of DOX was 1 mg/mL the optimal DOX loading efficiency was as high as 22.01%±0.43%. In vitro drug release behavior study demonstrated that HNTs/DOX/LIP shows a pH-responsive release property with fast drug release under acidic conditions (pH =5.4). MTT assays and in vivo experimental results revealed that HNTs/DOX/LIP exhibits a significantly higher inhibitory efficacy on the growth of mouse gastric cancer cells than free DOX at the same drug concentration. In addition, the life span of tumor-bearing mice in the HNTs/DOX/LIP-treated group was obviously prolonged compared with the control groups. Moreover, HNTs/DOX/LIP possessed excellent hemocompatibility as shown in the blood and histology studies. These findings indicated that the formed HNTs/DOX/LIP possesses higher antitumor efficacy and may be used as a targeted delivery nanoplatform for targeting therapy of different types of cancer cells.
Collapse
Affiliation(s)
- Kai Li
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yongxing Zhang
- Department of Orthopaedics, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Mengting Chen
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yangyang Hu
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Weiliang Jiang
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Li Zhou
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Sisi Li
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Min Xu
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Qinghua Zhao
- Department of Orthopaedics, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Rong Wan
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
43
|
Abdouh M, Hamam D, Gao ZH, Arena V, Arena M, Arena GO. Exosomes isolated from cancer patients' sera transfer malignant traits and confer the same phenotype of primary tumors to oncosuppressor-mutated cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:113. [PMID: 28854931 PMCID: PMC5577828 DOI: 10.1186/s13046-017-0587-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/24/2017] [Indexed: 12/14/2022]
Abstract
Background Horizontal transfer of malignant traits from the primary tumor to distant organs, through blood circulating factors, has recently become a thoroughly studied metastatic pathway to explain cancer dissemination. Recently, we reported that oncosuppressor gene-mutated human cells undergo malignant transformation when exposed to cancer patients’ sera. We also observed that oncosuppressor mutated cells would show an increased uptake of cancer-derived exosomes and we suggested that oncosuppressor genes might protect the integrity of the cell genome by blocking integration of cancer-derived exosomes. In the present study, we tested the hypothesis that cancer patients’ sera-derived exosomes might be responsible for the malignant transformation of target cells and that oncosuppressor mutation would promote their increased uptake. We also sought to unveil the mechanisms behind the hypothesized phenomena. Methods We used human BRCA1 knockout (BRCA1-KO) fibroblasts as target cells. Cells were treated in vitro with cancer patients’ sera or cancer patients’ sera-derived exosomes. Treated cells were injected into NOD-SCID mice. Immunohistochemical analyses were performed to determine the differentiation state of the xenotransplants. Mass spectrometry analyses of proteins from cancer exosomes and the BRCA1-KO fibroblasts’ membrane were performed to investigate possible de novo expression of molecules involved in vesicles uptake. Blocking of the identified molecules in vitro was performed and in vivo experiments were conducted to confirm the role of these molecules in the malignant transformation carried out by cancer-derived exosomes. Results Cells treated with exosomes isolated from cancer patients’ sera underwent malignant transformation and formed tumors when transplanted into immunodeficient mice. Histological analyses showed that the tumors were carcinomas that differentiated into the same lineage of the primary tumors of blood donors. Oncosuppressor mutation promoted the de novo expression, on the plasma membrane of target cells, of receptors, responsible for the increased uptake of cancer-derived exosomes. The selective blocking of these receptors inhibited the horizontal transfer of malignant traits. Conclusion These findings strengthen the hypothesis that oncogenic factors transferred via circulating cancer exosomes, induce malignant transformation of target cells even at distance. Oncosuppressor genes might protect the integrity of the cell genome by inhibiting the uptake of cancer-derived exosomes. Electronic supplementary material The online version of this article (10.1186/s13046-017-0587-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mohamed Abdouh
- Cancer Research Program, McGill University Health Centre-Research Institute, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada
| | - Dana Hamam
- Cancer Research Program, McGill University Health Centre-Research Institute, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada.,Department of Experimental Surgery, Faculty of Medicine, McGill University, 845 Rue Sherbrooke O, Montreal, Quebec, H3A 0G4, Canada
| | - Zu-Hua Gao
- Department of Pathology, McGill University Health Centre-Research Institute, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada
| | - Vincenzo Arena
- Department of Obstetrics and Gynecology, Santo Bambino Hospital, via Torre del Vescovo 4, Catania, Italy
| | - Manuel Arena
- Department of Surgical Sciences, Organ Transplantation and Advances Technologies, University of Catania, via Santa Sofia, 84, Catania, Italy
| | - Goffredo Orazio Arena
- Cancer Research Program, McGill University Health Centre-Research Institute, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada. .,Department of Surgery, McGill University, St. Mary Hospital, 3830 Lacombe Avenue, Montreal, Quebec, H3T 1M5, Canada.
| |
Collapse
|