1
|
Wu S, Nasser B Singab A, Lin G, Wang Y, Zhu H, Yang G, Chen J, Li J, Li P, Zhao D, Tian J, Ye L. The regulatory role of integrin in gastric cancer tumor microenvironment and drug resistance. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:130-136. [PMID: 39798809 DOI: 10.1016/j.pbiomolbio.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/18/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
Gastric cancer (GC) remains a significant global health burden due to its high aggressiveness, early metastasis, and poor prognosis. Despite advances in chemotherapy and targeted therapies, drug resistance remains a major obstacle to improving patient outcomes. Integrins, a family of transmembrane receptors, play a pivotal role in mediating tumor growth, invasion, and drug resistance by interacting with the tumor microenvironment (TME) and regulating signaling pathways such as Wnt/β-catenin, FAK, and MAPK. This review highlights the critical functions of various integrin subunits (e.g., α5, αv, β1, β3, β6) in promoting GC progression and their involvement in chemoresistance mechanisms. Additionally, integrins modulate immune cell infiltration and stromal cell interactions within the TME, further complicating GC treatment. Emerging evidence suggests that targeting integrins, either through inhibitors or integrin-specific therapeutic strategies, holds potential in overcoming drug resistance and improving clinical outcomes. This review underscores the need for further exploration of integrins as therapeutic targets in GC and suggests promising avenues for integrin-based therapies in personalized medicine.
Collapse
Affiliation(s)
- Songlin Wu
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Abdel Nasser B Singab
- Department of Pharmacognosy, Ain-Shams University, Cairo, Egypt; Centre of Drug Discovery Research and Development, Ain Shams University, Cairo, Egypt
| | - Guimei Lin
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; National Medical Products Administration Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong University, Jinan, Shandong, China
| | - Yulu Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Huaibo Zhu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Guang Yang
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Jiaqi Chen
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Jiaxuan Li
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Peiyao Li
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Di Zhao
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Jing Tian
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Lan Ye
- Center for Cancer Prevention and Treatment, Second Hospital of Shandong University, Jinan, Shandong, China.
| |
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
|
Zhang Z, Wang Z, Liu T, Tang J, Liu Y, Gou T, Chen K, Wang L, Zhang J, Yang Y, Zhang H. Exploring the role of ITGB6: fibrosis, cancer, and other diseases. Apoptosis 2024; 29:570-585. [PMID: 38127283 DOI: 10.1007/s10495-023-01921-6] [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] [Accepted: 11/07/2023] [Indexed: 12/23/2023]
Abstract
Integrin β6 (ITGB6), a member of the integrin family of proteins, is only present in epithelial tissues and frequently associates with integrin subunit αv to form transmembrane heterodimers named integrin αvβ6. Importantly, ITGB6 determines αvβ6 expression and availability. In addition to being engaged in organ fibrosis, ITGB6 is also directly linked to the emergence of cancer, periodontitis, and several potential genetic diseases. Therefore, it is of great significance to study the molecular-biological mechanism of ITGB6, which could provide novel insights for future clinical diagnosis and therapy. This review introduces the structure, distribution, and biological function of ITGB6. This review also expounds on ITGB6-related diseases, detailing the known biological effects of ITGB6.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, 627 Wuluo Road, Wuhan, 430070, China
| | - Tong Liu
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Jiayou Tang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Yanqing Liu
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Tiantian Gou
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Kangli Chen
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Li Wang
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Juan Zhang
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yang Yang
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Huan Zhang
- Department of Cardiology, Faculty of Life Sciences and Medicine, The Affiliated Hospital of Northwest University, Northwest University, Xi'an No.3 Hospital, Xi'an, 710021, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Faulty of Life Sciences and Medicine, Ministry of Education, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
4
|
Chen B, Liu J. Prospects and challenges of CAR-T in the treatment of ovarian cancer. Int Immunopharmacol 2024; 133:112112. [PMID: 38640714 DOI: 10.1016/j.intimp.2024.112112] [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: 03/12/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Ovarian cancer ranks as the seventh most prevalent cancer among women and is considered the most lethal gynecological malignancy on a global scale. The absence of reliable screening techniques, coupled with the insidious onset of nonspecific symptoms, often results in a delayed diagnosis, typically at an advanced stage characterized by peritoneal involvement. Management of advanced tumors typically involves a combination of chemotherapy and cytoreductive surgery. However, the therapeutic arsenal for ovarian cancer patients remains limited, highlighting the unmet need for precise, targeted, and sustained-release pharmacological agents. Genetically engineered T cells expressing chimeric antigen receptors (CARs) represent a promising novel therapeutic modality that selectively targets specific antigens, demonstrating robust and enduring antitumor responses in numerous patients. CAR T cell therapy has exhibited notable efficacy in hematological malignancies and is currently under investigation for its potential in treating various solid tumors, including ovarian cancer. Currently, numerous researchers are engaged in the development of novel CAR-T cells designed to target ovarian cancer, with subsequent evaluation of these candidate cells in preclinical studies. Given the ability of chimeric antigen receptor (CAR) expressing T cells to elicit potent and long-lasting anti-tumor effects, this therapeutic approach holds significant promise for the treatment of ovarian cancer. This review article examines the utilization of CAR-T cells in the context of ovarian cancer therapy.
Collapse
Affiliation(s)
- Biqing Chen
- Harbin Medical University, Harbin, Heilongjiang, China.
| | | |
Collapse
|
5
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024; 9:132. [PMID: 38763973 PMCID: PMC11102923 DOI: 10.1038/s41392-024-01823-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 05/21/2024] Open
Abstract
Tumor biomarkers, the substances which are produced by tumors or the body's responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Xinyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
6
|
Liao J, Chen R, Lin B, Deng R, Liang Y, Zeng J, Ma S, Qiu X. Cross-Talk between the TGF-β and Cell Adhesion Signaling Pathways in Cancer. Int J Med Sci 2024; 21:1307-1320. [PMID: 38818471 PMCID: PMC11134594 DOI: 10.7150/ijms.96274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024] Open
Abstract
Transforming growth factor-β (TGF-β) is strongly associated with the cell adhesion signaling pathway in cell differentiation, migration, etc. Mechanistically, TGF-β is secreted in an inactive form and localizes to the extracellular matrix (ECM) via the latent TGF-β binding protein (LTBP). However, it is the release of mature TGF-β that is essential for the activation of the TGF-β signaling pathway. This progress requires specific integrins (one of the main groups of cell adhesion molecules (CAMs)) to recognize and activate the dormant TGF-β. In addition, TGF-β regulates cell adhesion ability through modulating CAMs expression. The aberrant activation of the TGF-β signaling pathway, caused by abnormal expression of key regulatory molecules (such as Smad proteins, certain transcription factors, and non-coding RNAs), promotes tumor invasive and metastasis ability via epithelial-mesenchymal transition (EMT) during the late stages of tumorigenesis. In this paper, we summarize the crosstalk between TGF-β and cell adhesion signaling pathway in cancer and its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Jiahao Liao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Rentang Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Bihua Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Runhua Deng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Yanfang Liang
- Department of Pathology, Binhaiwan Central Hospital of Dongguan, Dongguan, Guangdong, 523905, China
| | - Jincheng Zeng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Sha Ma
- School of Biomedical Engineering, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Xianxiu Qiu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| |
Collapse
|
7
|
Urquiza M, Benavides-Rubio D, Jimenez-Camacho S. Structural analysis of peptide binding to integrins for cancer detection and treatment. Biophys Rev 2023; 15:699-708. [PMID: 37681100 PMCID: PMC10480133 DOI: 10.1007/s12551-023-01084-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/20/2023] [Indexed: 09/09/2023] Open
Abstract
Integrins are cell receptors involved in several metabolic pathways often associated with cell proliferation. Some of these integrins are downregulated during human physical development, but when these integrins are overexpressed in adult humans, they can be associated with several diseases, such as cancer. Molecules that specifically bind to these integrins are useful for cancer detection, diagnosis, and treatment. This review focuses on the structures of integrin-peptidic ligand complexes to dissect how the binding occurs and the molecular basis of the specificity and affinity of these peptidic ligands. Understanding these interactions at the molecular level is fundamental to be able to design new peptides that are more specific and more sensitive to a particular integrin. The integrin complexes covered in this review are α5β1, αIIbβ3, αvβ3, αvβ6, and αvβ8, because the molecular structures of the complex have been experimentally determined and their presence on tumor cancer cells are associated with a poor prognosis, making them targets for cancer detection and treatment.
Collapse
Affiliation(s)
- Mauricio Urquiza
- Chemistry Department, Faculty of Sciences, Universidad Nacional de Colombia, Carrera 30# 45-03, Ciudad Universitaria, Bogotá, Colombia
| | - Daniela Benavides-Rubio
- Chemistry Department, Faculty of Sciences, Universidad Nacional de Colombia, Carrera 30# 45-03, Ciudad Universitaria, Bogotá, Colombia
| | - Silvia Jimenez-Camacho
- Chemistry Department, Faculty of Sciences, Universidad Nacional de Colombia, Carrera 30# 45-03, Ciudad Universitaria, Bogotá, Colombia
| |
Collapse
|
8
|
Zhang B, Huang B, Zhang X, Li S, Zhu J, Chen X, Song H, Shang D. PANoptosis-related molecular subtype and prognostic model associated with the immune microenvironment and individualized therapy in pancreatic cancer. Front Oncol 2023; 13:1217654. [PMID: 37519797 PMCID: PMC10382139 DOI: 10.3389/fonc.2023.1217654] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/20/2023] [Indexed: 08/01/2023] Open
Abstract
Background PANoptosis is an inflammatory type of programmed cell death regulated by PANopotosome. Mounting evidence has shown that PANoptosis could be involved in cancer pathogenesis and the tumor immune microenvironment. Nevertheless, there have been no studies on the mechanism of PANoptosis on pancreatic cancer (PC) pathogenesis. Methods We downloaded the data on transcriptomic and clinical features of PC patients from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus databases. Additionally, the data on copy number variation (CNV), methylation and somatic mutations of genes in 33 types of cancers were obtained from TCGA. Next, we identified the PANoptosis-related molecular subtype using the consensus clustering analysis, and constructed and validated the PANoptosis-related prognostic model using LASSO and Cox regression analyses. Moreover, RT-qPCR was performed to determine the expression of genes involved in the model. Results We obtained 66 PANoptosis-related genes (PANRGs) from published studies. Of these, 24 PC-specific prognosis-related genes were identified. Pan-cancer analysis revealed complex genetic changes, including CNV, methylation, and mutation in PANRGs were identified in various cancers. By consensus clustering analysis, PC patients were classified into two PANoptosis-related patterns: PANcluster A and B. In PANcluster A, the patient prognosis was significantly worse compared to PANcluster B. The CIBERSORT algorithm showed a significant increase in the infiltration of CD8+ T cells, monocytes, and naïve B cells, in patients in PANcluster B. Additionally, the infiltration of macrophages, activated mast cells, and dendritic cells were higher in patients in PANcluster A. Patients in PANcluster A were more sensitive to erlotinib, selumetinib and trametinib, whereas patients in PANcluster B were highly sensitive to irinotecan, oxaliplatin and sorafenib. Moreover, we constructed and validated the PANoptosis-related prognostic model to predict the patient's survival. Finally, the GEPIA and Human Protein Atlas databases were analyzed, and RT-qPCR was performed. Compared to normal tissues, a significant increase in CXCL10 and ITGB6 (associated with the model) expression was observed in PC tissues. Conclusion We first identified the PANoptosis-related molecular subtypes and established a PANoptosis-related prognostic model for predicting the survival of patients with PC. These results would aid in exploring the mechanisms of PANoptosis in PC pathogenesis.
Collapse
Affiliation(s)
- Biao Zhang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bingqian Huang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xiaonan Zhang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Shuang Li
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jingyi Zhu
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xu Chen
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huiyi Song
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
9
|
Jiang M, Fang C, Ma Y. Prognosis Risk Model Based on Pyroptosis-Related lncRNAs for Gastric Cancer. Biomolecules 2023; 13:biom13030469. [PMID: 36979404 PMCID: PMC10046686 DOI: 10.3390/biom13030469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/29/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
Gastric cancer (GC) is a malignant tumor with a low survival rate, high recurrence rate, and poor prognosis. With respect to this, pyroptosis is a type of programmed cell death that can affect the occurrence and development of tumors. Indeed, long non-coding RNAs (lncRNAs) were broadly applied for the purposes of early diagnosis, treatment, and prognostic analysis in regard to cancer. Based on the association of these three purposes, we developed a novel prognosis risk model based on pyroptosis-related lncRNAs (PRlncRNAs) for GC. The PRlncRNAs were obtained via univariate and multivariate Cox regression in order to build the predictive signatures. The Kaplan–Meier and gene set enrichment analysis (GSEA) methods were used to evaluate the overall survival (OS) and functional differences between the high- and low-risk groups. Moreover, the correlation of the signatures with immune cell infiltration was determined through single-sample gene set enrichment analysis (ssGSEA). Finally, we analyzed this correlation with the treatment responses in the GC patients; then, we performed quantitative reverse transcription polymerase chain reactions (qRT-PCRs) in order to verify the risk model. The high-risk group received a worse performance in terms of prognosis and OS when compared to the low-risk group. With respect to this, the area under the receiver operating characteristic curve (ROC) was found to be 0.808. Through conducting the GSEA, it was found that the high-risk groups possessed a significant enrichment in terms of tumor–immunity pathways. Furthermore, the ssGSEA revealed that the predictive features possessed strong associations with immune cell infiltration in regard to GC. In addition, we highlighted that anti-immune checkpoint therapy, combined with conventional chemotherapy drugs, may be more suitable for high-risk patients. The expression levels of LINC01315, AP003392.1, AP000695.2, and HAGLR were significantly different between the GC cell lines and the normal cell lines. As such, the six PRlncRNAs could be regarded as important prognostic biomarkers for the purposes of subsequent diagnoses, treatments, prognostic predictions, and the mechanism research of GC.
Collapse
|
10
|
A novel metabolism-related prognostic gene development and validation in gastric cancer. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:447-459. [PMID: 36168087 DOI: 10.1007/s12094-022-02958-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/15/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND The importance of metabolism-related alterations in the development of gastric cancer (GC) is increasingly recognized. The present study aimed to identify metabolism-related genes to facilitate prognosis of GC patients. METHODS Gene expression datasets and clinical information of GC patients were downloaded from TCGA and GEO databases. We scored the enrichment of human metabolism-related pathways (n = 86) in GC samples by GSV, constructed prognostic risk models using LASSO algorithm and multivariate Cox regression analysis, combined with clinical information to construct a nomogram, and finally cis score algorithm to analyze the abundance of immune-related cells in different subtypes. We used Weka software to screen for prognosis-related marker genes and finally validated the expression of the selected genes in clinical cancer patient tissues. RESULTS We identified that two GC metabolism-related signatures were strongly associated with OS and the levels of immune cell infiltration. Moreover, a survival prediction model for GC was established based on six GC metabolism-related genes. Time-dependent ROC analysis showed good stability of the risk prediction scoring model. The model was successfully validated in an independent ACRG cohort, and the expression trends of key genes were also verified in the GC tissues of patients. DLX1, LTBP2, FGFR1 and MMP2 were highly expressed in the cluster with poorer prognosis while SLC13A2 and SLCO1B3 were highly expressed in the cluster with better prognosis. CONCLUSIONS We identified a risk predictive score model based on six metabolism-related genes related to survival, which may serve as prognostic indicators and potential therapeutic targets for GC.
Collapse
|
11
|
Matsumoto Y, Kage H, Morota M, Zokumasu K, Ando T, Maemura K, Watanabe K, Kawakami M, Hinata M, Ushiku T, Nakajima J, Nagase T. Integrin alpha 2 is associated with tumor progression and postoperative recurrence in non-small cell lung cancer. Jpn J Clin Oncol 2023; 53:63-73. [PMID: 36151049 DOI: 10.1093/jjco/hyac148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 08/29/2022] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Integrins are transmembrane proteins that mediate cell adhesion to extracellular matrix. Whereas expression of integrin alpha 2 is associated with motility, invasiveness and cellular differentiation in various tumors, the role of integrin alpha 2 in lung cancer has not been studied in detail. The aim of this study was to determine whether and how aberrant integrin alpha 2 expression in non-small cell lung cancer leads to different outcomes. METHODS We measured expression of integrin alpha 2 by quantitative polymerase chain reaction in 100 samples collected from non-small cell lung cancer patients who had undergone surgical resection. We assigned patients to high and low expression groups and analyzed survival. Cellular morphology, adhesion, proliferation, migration and invasion were examined in human lung cancer cell lines. RESULTS Among 100 cases, 41 were female, with a median age of 71 years. High expression of integrin alpha 2 in non-small cell lung cancer was associated with lower recurrence-free survival (P = 0.004). Overexpression of integrin alpha 2 in cell lines had no effect on cell proliferation or invasion but resulted in increased cell size (1416 μm2 versus 470 μm2 in H522 cells, P < 0.001; 1822 μm2 versus 1029 μm2 in H661 cells, P = 0.02), adhesion (P < 0.001 in H522 and H661 cells) and migration (gap area filled was 71% versus 36% in H522 cells, P < 0.001; 57% versus 26% in H661 cells, P = 0.001). These changes were suppressed by E7820, an inhibitor of integrin alpha 2. CONCLUSIONS Integrin alpha 2 may play a significant role in lung cancer adhesion and migration, and may lead to a higher risk of recurrence.
Collapse
Affiliation(s)
- Yoko Matsumoto
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Hidenori Kage
- Next-Generation Precision Medicine Development Laboratory, The University of Tokyo, Tokyo, Japan
| | - Mizuki Morota
- Department of Thoracic Surgery, The University of Tokyo, Tokyo, Japan
| | - Koichi Zokumasu
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahiro Ando
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Keita Maemura
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Kousuke Watanabe
- Department of Clinical Laboratory, The University of Tokyo, Tokyo, Japan
| | - Masanori Kawakami
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Tetsuo Ushiku
- Department of Pathology, The University of Tokyo, Tokyo, Japan
| | - Jun Nakajima
- Department of Thoracic Surgery, The University of Tokyo, Tokyo, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Integrin Alpha v Beta 6 (αvβ6) and Its Implications in Cancer Treatment. Int J Mol Sci 2022; 23:ijms232012346. [PMID: 36293202 PMCID: PMC9603893 DOI: 10.3390/ijms232012346] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/20/2022] Open
Abstract
Integrins are necessary for cell adhesion, migration, and positioning. Essential for inducing signalling events for cell survival, proliferation, and differentiation, they also trigger a variety of signal transduction pathways involved in mediating invasion, metastasis, and squamous-cell carcinoma. Several recent studies have demonstrated that the up- and down-regulation of the expression of αv and other integrins can be a potent marker of malignant diseases and patient prognosis. This review focuses on an arginine-glycine-aspartic acid (RGD)-dependent integrin αVβ6, its biology, and its role in healthy humans. We examine the implications of αVβ6 in cancer progression and the promotion of epithelial-mesenchymal transition (EMT) by contributing to the activation of transforming growth factor beta TGF-β. Although αvβ6 is crucial for proper function in healthy people, it has also been validated as a target for cancer treatment. This review briefly considers aspects of targeting αVβ6 in the clinic via different therapeutic modalities.
Collapse
|
13
|
Liu D, Liu S, Fang Y, Liu L, Hu K. Comprehensive Analysis of the Expression and Prognosis for ITGBs: Identification of ITGB5 as a Biomarker of Poor Prognosis and Correlated with Immune Infiltrates in Gastric Cancer. Front Cell Dev Biol 2022; 9:816230. [PMID: 35223869 PMCID: PMC8863963 DOI: 10.3389/fcell.2021.816230] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/27/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Integrin β superfamily members (ITGBs) are documented to play important roles in various biological processes, and accumulating evidence suggests that ITGBs are associated with carcinogenic effects in several malignancies. Gastric cancer (GC) is a complicated and highly heterogeneous disease; however, the expression and prognostic values of eight ITGBs and potential mechanism in GC remain largely unclear. Methods: The expression and prognostic significance of ITGBs in GC were systematically analyzed through Gene Expression Profiling Interactive Analysis, Human Protein Atlas, Kaplan–Meier Plotter, and cBioPortal databases. Then, the mRNA transcription data and corresponding clinical data of GC were downloaded from the Gene Expression Omnibus database as a testing cohort, and differentially expressed and prognostic genes were identified. The correlation between ITGB5 expression and overall survival and various clinical parameters were found by using univariate/multivariable Cox regression and Kaplan–Meier survival analysis. Additionally, differential analysis of gene expression profiles in low- and high-ITGB5 expression groups and pathway enrichment analysis was performed. Finally, the correlation of ITGB5 expression with immune infiltrates in GC was clarified. Results: Compared with adjacent normal tissue, the results reveal that the mRNA levels of ITGB1-2 and ITGB4-8 are significantly higher in GC, and immunohistochemistry results show the consistency between RNA and protein expression levels. Cox regression and Kaplan–Meier survival analysis indicate that high ITGB5 expression contributes to a poor prognosis and could be an independent prognostic factor in GC patients. Besides this, gene functional enrichment analysis indicates that ITGB5 expression is significantly associated with extracellular matrix organization, cell-substrate adhesion, and ossification. The KEGG pathway analysis of ITGB5 shows a close association between ITGB5 and focal adhesion, ECM-receptor interaction, phagosome, and PI3K-Akt signaling pathway. Last, the infiltrating level of CD4+ T cells, macrophages, and dendritic cells are positively related to the expression of ITGB5, especially macrophages, and lower levels of macrophages predict a better prognosis in GC in our study. Conclusion: Our findings investigate that ITGB5 may function as a valid biomarker of prognosis, and high expression of ITGB5 predicts poor prognosis for patients with GC. Besides this, it might be a potential target of precision therapy against GC.
Collapse
Affiliation(s)
- Dongliang Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shaojun Liu
- Department of General Surgery, The First Hospital Affiliated to the University of Science and Technology of China, Hefei, China
| | - Yu Fang
- Department of General Surgery, The First Hospital Affiliated to the University of Science and Technology of China, Hefei, China
| | - Liu Liu
- Department of General Surgery, The First Hospital Affiliated to the University of Science and Technology of China, Hefei, China
- *Correspondence: Liu Liu, ; Kongwang Hu,
| | - Kongwang Hu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Liu Liu, ; Kongwang Hu,
| |
Collapse
|
14
|
Li Z, Sun Y, Xu J, Yang H, Liu X, Tian Y, Cao S, Zhou Y. Integrin-β6 Serves as a Potential Prognostic Serum Biomarker for Gastric Cancer. Front Oncol 2021; 11:770997. [PMID: 34796117 PMCID: PMC8593195 DOI: 10.3389/fonc.2021.770997] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/08/2021] [Indexed: 12/25/2022] Open
Abstract
Discovering novel biomarkers that easily accessed is a key step towards the personalized medicine approach for gastric cancer patients. Integrin-β6 (ITGB6) is a subtype of integrin that is exclusively expressed on the surface of epithelial cells and is up-regulated in various tumors. In the present study, a retrospective cohort with 135 gastric cancer patients and a prospective cohort with 34 gastric cancer patients were constructed, ITGB6 expression were detected in both the serum specimens and the tissue specimens. Detailed clinicopathological parameters as well as patients' survival were recorded. A nomogram including ITGB6 expression was also constructed and validated to predict the prognosis of gastric cancer patients. Results showed that serum ITGB6 expression was obviously increased and associated with tumor stage in gastric cancer patients, serum ITGB6 expression was relatively high in patients with liver metastasis. High ITGB6 expression indicated a poor prognosis, and nomogram including serum ITGB6 expression could predict the prognosis of gastric cancer patients effectively. Moreover, serum ITGB6 expression was associated with ITGB6 expression in tumor tissues. Furthermore, combined serum ITGB6 and CEA levels contributed to the risk stratification and prognostic prediction for gastric cancer patients. In addition, the serum expression of ITGB6 decreased significantly after radical surgery, and a new rise in serum ITGB6 expression indicated tumor recurrence or progression. The present study identified a novel serum biomarker for the risk stratification, prognostic prediction and surveillance of gastric cancer patients.
Collapse
Affiliation(s)
- Zequn Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuqi Sun
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianfei Xu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hao Yang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaodong Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yulong Tian
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shougen Cao
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanbing Zhou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
15
|
Molecular Targets for Gastric Cancer Treatment and Future Perspectives from a Clinical and Translational Point of View. Cancers (Basel) 2021; 13:cancers13205216. [PMID: 34680363 PMCID: PMC8533881 DOI: 10.3390/cancers13205216] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer is a leading cause of cancer death worldwide. Systemic treatment comprising chemotherapy and targeted therapy is the standard of care in advanced/metastatic gastric cancer. Comprehensive molecular characterization of gastric adenocarcinomas by the TCGA Consortium and ACRG has resulted in the definition of distinct molecular subtypes. These efforts have in parallel built a basis for the development of novel molecularly stratified treatment approaches. Based on this molecular characterization, an increasing number of specific genomic alterations can potentially serve as treatment targets. Consequently, the development of promising compounds is ongoing. In this review, key molecular alterations in gastric and gastroesophageal junction cancers will be addressed. Finally, the current status of the translation of targeted therapy towards clinical applications will be reviewed.
Collapse
|
16
|
Wang H, Zhang M, Zhang Y, Liu Y, Wang M, Liu Y, Liao Y, Li Z, Feng Y, Chen J. The decreased expression of integrin αv is involved in T-2 toxin-induced extracellular matrix degradation in chondrocytes. Toxicon 2021; 199:109-116. [PMID: 34139256 DOI: 10.1016/j.toxicon.2021.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/18/2021] [Accepted: 06/07/2021] [Indexed: 01/10/2023]
Abstract
T-2 toxin is one of the most toxic and common mycotoxins in grains and related products. It is considered a risk factor for Kashin-Beck disease (KBD), an endemic osteoarthritis. Both in vitro and in vivo studies have shown that T-2 toxin can cause extracellular matrix degradation; however, the underlying mechanism is unclear. Integrins have been found to regulate the expression of matrix metalloproteinases (MMPs), the 'scissors' of matrix proteins. In this study, we investigated whether integrin αv played a role in T-2 toxin-induced matrix degradation. Results from our study showed that the expression of integrin αv in the cartilage of rats fed T-2 toxin was reduced compared to that in rats fed a normal diet. Integrin αv was downregulated in T-2 toxin-treated C28/I2 chondrocytes, and selenium was found to have a protective effect. The expression of MMP-1, -3, -10, and -13 increased whereas that of type II collagen (Col II) protein decreased in C28/I2 cells treated with an integrin αv inhibitor. In conclusion, T-2 toxin can downregulate integrin αv expression in chondrocytes. Reduced integrin αv signalling could induce the release of MMPs, leading to matrix degradation.
Collapse
Affiliation(s)
- Hui Wang
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Meng Zhang
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Ying Zhang
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Yinan Liu
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Mengying Wang
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Yue Liu
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Yucheng Liao
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Zhengzheng Li
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Yiping Feng
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Jinghong Chen
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China.
| |
Collapse
|
17
|
Desnoyers A, González C, Pérez-Segura P, Pandiella A, Amir E, Ocaña A. Integrin ανβ6 Protein Expression and Prognosis in Solid Tumors: A Meta-Analysis. Mol Diagn Ther 2021; 24:143-151. [PMID: 32100239 DOI: 10.1007/s40291-020-00450-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND OBJECTIVE Integrins are a family of adhesion receptor proteins that provide signaling from the extracellular matrix to the cytoplasm. They have been associated with cancer by promoting migration, invasion, metastasis, and survival. ανβ6 integrin is upregulated in several tumors. Here, we evaluate the prognostic impact of ανβ6 integrin protein expression in solid tumors. METHODS A systematic search of electronic databases identified publications exploring the effect of ανβ6 integrin on overall survival (OS). Hazard ratios (HRs) were pooled in a meta-analysis using generic inverse variance and random effects modeling. Subgroup analyses were conducted based on tumor site, tumor stage, antibody used for immunohistochemistry (IHC) and method for extraction of the HR. A meta-regression explored the influence of clinical variables on the magnitude of effect of ανβ6 integrins on OS. RESULTS Seventeen studies comprising 5795 patients met the inclusion criteria. High ανβ6 integrin expression in tumors was associated with worse OS (HR 1.65, 95% confidence interval [CI] 1.32-2.06; Cochran's Q p < 0.001, I2 = 81%). Adverse outcomes were similar in all tumor sites (subgroup difference p = 0.10), with the strongest association between ανβ6 integrins and OS in gastric cancer (HR 2.20, 95% CI 1.71-2.83) and the lowest in head and neck cancer (HR 1.21, 95% CI 0.79-1.83). There was no significant difference between early-stage and metastatic cancer, type of IHC antibodies, and analysis methods. CONCLUSIONS High expression of ανβ6 integrins is associated with adverse survival outcome in several tumors. Prospective studies evaluating the prognostic impact of ανβ6 integrin and its role as a therapeutic target are warranted.
Collapse
Affiliation(s)
- Alexandra Desnoyers
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, The University of Toronto, Toronto, ON, Canada
| | - Carlos González
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, The University of Toronto, Toronto, ON, Canada.,Experimental Therapeutics Unit, Medical Oncology Department, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain.,Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain.,CIC-Universidad de Salamanca, Salamanca, Spain.,Centro Regional de Investigaciones Biomédicas, Castilla-La Mancha University (UCLM), Albacete, Spain
| | - Pedro Pérez-Segura
- Experimental Therapeutics Unit, Medical Oncology Department, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Atanasio Pandiella
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain.,CIC-Universidad de Salamanca, Salamanca, Spain
| | - Eitan Amir
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, The University of Toronto, Toronto, ON, Canada
| | - Alberto Ocaña
- Experimental Therapeutics Unit, Medical Oncology Department, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain. .,Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain. .,Centro Regional de Investigaciones Biomédicas, Castilla-La Mancha University (UCLM), Albacete, Spain.
| |
Collapse
|
18
|
Shan ZG, Sun ZW, Zhao LQ, Gou Q, Chen ZF, Zhang JY, Chen W, Su CY, You N, Zhuang Y, Zhao YL. Upregulation of Tubulointerstitial nephritis antigen like 1 promotes gastric cancer growth and metastasis by regulating multiple matrix metallopeptidase expression. J Gastroenterol Hepatol 2021; 36:196-203. [PMID: 32537806 DOI: 10.1111/jgh.15150] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/27/2020] [Accepted: 06/11/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIM Tubulointerstitial nephritis antigen-like 1 (TINAGL1), as a novel matricellular protein, has been demonstrated to participate in cancer progression, whereas the potential function of TINAGL1 in gastric cancer (GC) remains unknown. METHODS The expression pattern of TINAGL1 in GC was examined by immunohistochemistry, ELISA, real-time polymerase chain reaction, and Western blot. Correlation between TINAGL1 and matrix metalloproteinases (MMPs) was analyzed by the GEPIA website and Kaplan-Meier plots database. The lentivirus-based TINAGL1 knockdown, CCK-8, and transwell assays were used to test the function of TINAGL1 in vitro. The role of TINAGL1 was confirmed by subcutaneous xenograft, abdominal dissemination, and lung metastasis model. Microarray experiments, ELISA, real-time polymerase chain reaction, and Western blot were used to identify molecular mechanism. RESULTS TINAGL1 was increased in GC tumor tissues and associated with poor patient survival. Moreover, TINAGL1 significantly promoted GC cell proliferation and migration in vitro as well as facilitated GC tumor growth and metastasis in vivo. TINAGL1 expression in GC cells was accompanied with increasing MMPs including MMP2, MMP9, MMP11, MMP14, and MMP16. GEPIA database revealed that these MMPs were correlated with TINAGL1 in GC tumors and that the most highly expressed MMP was MMP2. Mechanically, TINAGL1 regulated MMP2 through the JNK signaling pathway activation. CONCLUSIONS Our data highlight that TINAGL1 promotes GC growth and metastasis and regulates MMP2 expression, indicating that TINAGL1 may serve as a therapeutic target for GC.
Collapse
Affiliation(s)
- Zhi-Guo Shan
- Department of General Surgery and Centre of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhen-Wei Sun
- The 988 Hospital of PLA, Zhengzhou, Henan, China
| | - Li-Qun Zhao
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Qiang Gou
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Zhi-Fu Chen
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Jin-Yu Zhang
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Weisan Chen
- La Trobe Institute of Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Chong-Yu Su
- Department of General Surgery and Centre of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Nan You
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuan Zhuang
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Yong-Liang Zhao
- Department of General Surgery and Centre of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
19
|
Chen C, Ge X, Zhao Y, Wang D, Ling L, Zheng S, Ding K, Wang J, Sun L. Molecular Alterations in Metastatic Ovarian Cancer From Gastrointestinal Cancer. Front Oncol 2020; 10:605349. [PMID: 33363035 PMCID: PMC7758447 DOI: 10.3389/fonc.2020.605349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/03/2020] [Indexed: 12/24/2022] Open
Abstract
Reports indicate that most metastatic ovarian cancer (MOC) originates from gastrointestinal cancer (GIC). Notably, GICs metastasize to the ovary frequently via 3 main routes including hematogenous spread, lymphogenous spread, and transcoelomic spread. Nonetheless, the mechanism of the progression remains unknown, and only a handful of literature exists on the molecular alteration implicated in MOC from GIC. This work collected existing evidence and literature on the vital molecules of the metastatic pathway and systematically analyzed them geared toward exploring the mechanism of the metastatic pathway of MOC. Further, this review described dominating molecular alteration in the metastatic process from cancer cells detaching away from lesions to arrive at the ovary, including factors for regulating signaling pathways in epithelial-interstitial transformation, invading, and surviving in the circulatory system or abdominal cavity. We interrogated the basis of the ovary as a distant metastatic site. This article provides new insights into the metastatic pathway and generates novel therapeutic targets for effective treatment and satisfactory outcomes in GIC patients.
Collapse
Affiliation(s)
- Chao Chen
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxu Ge
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yamei Zhao
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Da Wang
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Limian Ling
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Zheng
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Wang
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lifeng Sun
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
20
|
Urquiza M, Guevara V, Diaz-Sana E, Mora F. The Role of αvβ6 Integrin Binding Molecules in the Diagnosis and Treatment of Cancer. CURR ORG CHEM 2020. [DOI: 10.2174/1385272824999200528124936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Peptidic and non-peptidic αvβ6 integrin-binding molecules have been used in
the clinic for detection and treatment of tumors expressing αvβ6 integrin, because this protein
is expressed in malignant epithelial cells of the oral cavity, pancreas, breast, ovary,
colon and stomach carcinomas but it is not expressed in healthy adult tissue except during
wound healing and inflammation. This review focuses on the landscape of αvβ6 integrinbinding
molecules and their use in cancer treatment and detection, and discusses recent
designs for tumor detection, treatment, and immunotherapy. In the last ten years, several
reviews abamp;#945;vβ6 integrin-binding molecules and their role in cancer detection and treatment.
Firstly, this review describes the role of the αvβ6 integrin in normal tissues, how the expression
of this protein is correlated with cancer severity and its role in cancer development. Taking into account
the potential of αvβ6 integrin-binding molecules in detection and treatment of specific tumors, special
attention is given to several high-affinity αvβ6 integrin-binding peptides used for tumor imaging; particularly,
the αvβ6-binding peptide NAVPNLRGDLQVLAQKVART [A20FMDV2], derived from the foot and mouth
disease virus. This peptide labeled with either 18F, 111In or with 68Ga has been used for PET imaging of αvβ6
integrin-positive tumors. Moreover, αvβ6 integrin-binding peptides have been used for photoacoustic and fluorescence
imaging and could potentially be used in clinical application in cancer diagnosis and intraoperative
imaging of αvβ6-integrin positive tumors. Additionally, non-peptidic αvβ6-binding molecules have been designed
and used in the clinic for the detection and treatment of αvβ6-expressing tumors. Anti-αvβ6 integrin antibodies
are another useful tool for selective identification and treatment of αvβ6 (+) tumors. The utility of
these αvβ6 integrin-binding molecules as a tool for tumor detection and treatment is discussed, considering
specificity, sensitivity and serum stability. Another use of the αvβ6 integrin-binding peptides is to modify the
Ad5 cell tropism for inducing oncolytic activity of αvβ6-integrin positive tumor cells by expressing
A20FMDV2 peptide within the fiber knob protein (Ad5NULL-A20). The newly designed oncolytic
Ad5NULL-A20 virotherapy is promising for local and systemic targeting of αvβ6-overexpressing cancers. Finally,
new evidence has emerged, indicating that chimeric antigen receptor (CAR) containing the αvβ6 integrin-
binding peptide on top of CD28+CD3 endodomain displays a potent therapeutic activity in a diverse
repertoire of solid tumor models.
Collapse
Affiliation(s)
- Mauricio Urquiza
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| | - Valentina Guevara
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| | - Erika Diaz-Sana
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| | - Felipe Mora
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| |
Collapse
|
21
|
Sun Q, Dong X, Shang Y, Sun F, Niu J, Li F. Integrin αvβ6 predicts poor prognosis and promotes resistance to cisplatin in hilar cholangiocarcinoma. Pathol Res Pract 2020; 216:153022. [PMID: 32534716 DOI: 10.1016/j.prp.2020.153022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/23/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Integrin αvβ6 is associated with an extremely aggressive cancer phenotype. However, little is known about the clinicopathological significance and prognostic value of integrin αvβ6 in human hilar cholangiocarcinoma. METHODS In the present study, bioinformatics analysis demonstrated a significant increase of integrin β6 gene expression in cholangiocarcinoma tissues compared to non-tumorous tissues, which was further validated in clinical samples through RT-qPCR and western blotting analyses. Integrin αvβ6 was observed to be expressed in 48.6% of tumors, and its expression was related to a poor tumor differentiation (p = 0.002), lymph node metastasis (p<0.001) and advanced TNM stage (p=0.001). Furthermore, patients who were αvβ6-positive showed a significantly shorter overall survival period than those who were αvβ6-negative (p=0.004). Multivariate analysis confirmed that integrin αvβ6 was an independent prognostic factor (p=0.002). In addition, loss- and gain-of-function assays showed integrin αvβ6 not only played an important role in colony formation, but also protected cholangiocarcinoma cells from cisplatin-induced growth inhibition and apoptosis. ERK/MAPK signaling pathway was involved in integrin αvβ6-mediated resistance of cholangiocarcinoma cells to cisplatin. CONCLUSIONS Taken together, the present findings revealed that integrin αvβ6 could serve as a potential prognostic predictor and contribute to cisplatin resistance, which might prove to be a promising target candidate for the clinical intervention of human hilar cholangiocarcinoma.
Collapse
Affiliation(s)
- Qi Sun
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiwen Dong
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yukui Shang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Fengkai Sun
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Jun Niu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Fanni Li
- Department of Talent Highland, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
22
|
The Extracellular Matrix: An Accomplice in Gastric Cancer Development and Progression. Cells 2020; 9:cells9020394. [PMID: 32046329 PMCID: PMC7072625 DOI: 10.3390/cells9020394] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) is a dynamic and highly organized tissue structure, providing support and maintaining normal epithelial architecture. In the last decade, increasing evidence has emerged demonstrating that alterations in ECM composition and assembly strongly affect cellular function and behavior. Even though the detailed mechanisms underlying cell-ECM crosstalk are yet to unravel, it is well established that ECM deregulation accompanies the development of many pathological conditions, such as gastric cancer. Notably, gastric cancer remains a worldwide concern, representing the third most frequent cause of cancer-associated deaths. Despite increased surveillance protocols, patients are usually diagnosed at advanced disease stages, urging the identification of novel diagnostic biomarkers and efficient therapeutic strategies. In this review, we provide a comprehensive overview regarding expression patterns of ECM components and cognate receptors described in normal gastric epithelium, pre-malignant lesions, and gastric carcinomas. Important insights are also discussed for the use of ECM-associated molecules as predictive biomarkers of the disease or as potential targets in gastric cancer.
Collapse
|
23
|
FARP1 boosts CDC42 activity from integrin αvβ5 signaling and correlates with poor prognosis of advanced gastric cancer. Oncogenesis 2020; 9:13. [PMID: 32029704 PMCID: PMC7005035 DOI: 10.1038/s41389-020-0190-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 12/11/2019] [Accepted: 01/10/2020] [Indexed: 02/01/2023] Open
Abstract
Considering the poor prognosis of most advanced cancers, prevention of invasion and metastasis is essential for disease control. Ras homologous (Rho) guanine exchange factors (GEFs) and their signaling cascade could be potential therapeutic targets in advanced cancers. We conducted in silico analyses of The Cancer Genome Atlas expression data to identify candidate Rho-GEF genes showing aberrant expression in advanced gastric cancer and found FERM, Rho/ArhGEF, and pleckstrin domain protein 1 (FARP1) expression is related to poor prognosis. Analyses in 91 clinical advanced gastric cancers of the relationship of prognosis and pathological factors with immunohistochemical expression of FARP1 indicated that high expression of FARP1 is significantly associated with lymphatic invasion, lymph metastasis, and poor prognosis of the patients (P = 0.025). In gastric cancer cells, FARP1 knockdown decreased cell motility, whereas FARP1 overexpression promoted cell motility and filopodium formation via CDC42 activation. FARP1 interacted with integrin β5, and a potent integrin αvβ5 inhibitor (SB273005) prevented cell motility in only high FARP1-expressing gastric cancer cells. These results suggest that the integrin αvβ5-FARP1-CDC42 axis plays a crucial role in gastric cancer cell migration and invasion. Thus, regulatory cascade upstream of Rho can be a specific and promising target of advanced cancer treatment.
Collapse
|
24
|
Rajabi M, Adeyeye M, Mousa SA. Peptide-Conjugated Nanoparticles as Targeted Anti-angiogenesis Therapeutic and Diagnostic in Cancer. Curr Med Chem 2019; 26:5664-5683. [DOI: 10.2174/0929867326666190620100800] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/11/2019] [Accepted: 03/21/2019] [Indexed: 12/25/2022]
Abstract
:Targeting angiogenesis in the microenvironment of a tumor can enable suppression of tumor angiogenesis and delivery of anticancer drugs into the tumor. Anti-angiogenesis targeted delivery systems utilizing passive targeting such as Enhanced Permeability and Retention (EPR) and specific receptor-mediated targeting (active targeting) should result in tumor-specific targeting. One targeted anti-angiogenesis approach uses peptides conjugated to nanoparticles, which can be loaded with anticancer agents. Anti-angiogenesis agents can suppress tumor angiogenesis and thereby affect tumor growth progression (tumor growth arrest), which may be further reduced with the targetdelivered anticancer agent. This review provides an update of tumor vascular targeting for therapeutic and diagnostic applications, with conventional or long-circulating nanoparticles decorated with peptides that target neovascularization (anti-angiogenesis) in the tumor microenvironment.
Collapse
Affiliation(s)
- Mehdi Rajabi
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| | - Mary Adeyeye
- Department of Chemistry, University of Albany, State University of New York, Albany, NY 12222, United States
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| |
Collapse
|
25
|
Łasiñska I, Mackiewicz J. Integrins as A New Target for Cancer Treatment. Anticancer Agents Med Chem 2019; 19:580-586. [DOI: 10.2174/1871520618666181119103413] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/16/2018] [Accepted: 11/10/2018] [Indexed: 12/19/2022]
Abstract
:Despite the great progress in the development of targeted therapies for different types of cancer utilizing monoclonal antibodies (e.g., cetuximab for colorectal cancer and head and neck cancer therapy), kinase inhibitors (e.g., sorafenib for kidney cancer and gastrointestinal stromal tumours therapy), and immunomodulatory treatments (e.g., nivolumab and pembrolizumab for melanoma therapy and lung cancer therapy), there is still a need to search for new, more effective treatments.:Integrins are responsible for intercellular adhesion and interaction with the cellular matrix. The function of integrins is related to the transduction of intracellular signals associated with adhesion, migration, cell proliferation, differentiation, and apoptosis. Molecules targeting integrins that lead to cancer cell death have been developed. The most advanced molecules studied in clinical trials are abituzumab, intetumumab and cilengitide. There are different groups of anti-integrin drugs: monoclonal antibodies (e.g., abituzumab) and other such as cilengitide, E7820 and MK-0429. These drugs have been evaluated in various cancer types. However, they have shown modest efficacy, and none of them have yet been approved for cancer treatment. Studies have shown that patient selection using biomarkers might improve the efficacy of anti-integrin cancer treatment. Many preclinical models have demonstrated promising results using integrin visualization for cancer detection and treatment efficacy monitoring; however, these strategies require further evaluation in humans.
Collapse
Affiliation(s)
- Izabela Łasiñska
- Department of Medical and Experimental Oncology, Heliodor Swiecicki University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| | - Jacek Mackiewicz
- Department of Medical and Experimental Oncology, Heliodor Swiecicki University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
26
|
Whilding LM, Halim L, Draper B, Parente-Pereira AC, Zabinski T, Davies DM, Maher J. CAR T-Cells Targeting the Integrin αvβ6 and Co-Expressing the Chemokine Receptor CXCR2 Demonstrate Enhanced Homing and Efficacy against Several Solid Malignancies. Cancers (Basel) 2019; 11:E674. [PMID: 31091832 PMCID: PMC6563120 DOI: 10.3390/cancers11050674] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/07/2019] [Accepted: 05/10/2019] [Indexed: 02/06/2023] Open
Abstract
Despite the unprecedented clinical success of chimeric antigen receptors (CAR) T-cells against haematological malignancy, solid tumors impose a far greater challenge to success. Largely, this stems from an inadequate capacity of CAR T-cells that can traffic and maintain function within a hostile microenvironment. To enhance tumor-directed T-cell trafficking, we have engineered CAR T-cells to acquire heightened responsiveness to interleukin (IL)-8. Circulating IL-8 levels correlate with disease burden and prognosis in multiple solid tumors in which it exerts diverse pathological functions including angiogenesis, support of cancer stem cell survival, and recruitment of immunosuppressive myeloid cells. To harness tumor-derived IL-8 for therapeutic benefit, we have co-expressed either of its cognate receptors (CXCR1 or CXCR2) in CAR T-cells that target the tumor-associated αvβ6 integrin. We demonstrate here that CXCR2-expressing CAR T-cells migrate more efficiently towards IL-8 and towards tumor conditioned media that contains this cytokine. As a result, these CAR T-cells elicit superior anti-tumor activity against established αvβ6-expressing ovarian or pancreatic tumor xenografts, with a more favorable toxicity profile. These data support the further engineering of CAR T-cells to acquire responsiveness to cancer-derived chemokines in order to improve their therapeutic activity against solid tumors.
Collapse
Affiliation(s)
- Lynsey M Whilding
- King's College London, School of Cancer and Pharmaceutical Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
| | - Leena Halim
- King's College London, School of Cancer and Pharmaceutical Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
| | - Benjamin Draper
- King's College London, School of Cancer and Pharmaceutical Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
| | - Ana C Parente-Pereira
- King's College London, School of Cancer and Pharmaceutical Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
| | - Tomasz Zabinski
- King's College London, School of Cancer and Pharmaceutical Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
| | - David Marc Davies
- King's College London, School of Cancer and Pharmaceutical Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
| | - John Maher
- King's College London, School of Cancer and Pharmaceutical Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London SE5 9RS, UK.
- Department of Immunology, Eastbourne Hospital, East Sussex BN21 2UD, UK.
| |
Collapse
|
27
|
Arun AS, Tepper CG, Lam KS. Identification of integrin drug targets for 17 solid tumor types. Oncotarget 2018; 9:30146-30162. [PMID: 30046394 PMCID: PMC6059022 DOI: 10.18632/oncotarget.25731] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
Integrins are contributors to remodeling of the extracellular matrix and cell migration. Integrins participate in the assembly of the actin cytoskeleton, regulate growth factor signaling pathways, cell proliferation, and control cell motility. In solid tumors, integrins are involved in promoting metastasis to distant sites, and angiogenesis. Integrins are a key target in cancer therapy and imaging. Integrin antagonists have proven successful in halting invasion and migration of tumors. Overexpressed integrins are prime anti-cancer drug targets. To streamline the development of specific integrin cancer therapeutics, we curated data to predict which integrin heterodimers are pausible therapeutic targets against 17 different solid tumors. Computational analysis of The Cancer Genome Atlas (TCGA) gene expression data revealed a set of integrin targets that are differentially expressed in tumors. Filtered by FPKM (Fragments Per Kilobase of transcript per Million mapped reads) expression level, overexpressed subunits were paired into heterodimeric protein targets. By comparing the RNA-seq differential expression results with immunohistochemistry (IHC) data, overexpressed integrin subunits were validated. Biologics and small molecule drug compounds against these identified overexpressed subunits and heterodimeric receptors are potential therapeutics against these cancers. In addition, high-affinity and high-specificity ligands against these integrins can serve as efficient vehicles for delivery of cancer drugs, nanotherapeutics, or imaging probes against cancer.
Collapse
Affiliation(s)
- Adith S Arun
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, UC Davis NCI-Designated Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Clifford G Tepper
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, UC Davis NCI-Designated Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, UC Davis NCI-Designated Comprehensive Cancer Center, Sacramento, CA 95817, USA
| |
Collapse
|
28
|
Molaei F, Forghanifard MM, Fahim Y, Abbaszadegan MR. Molecular Signaling in Tumorigenesis of Gastric Cancer. IRANIAN BIOMEDICAL JOURNAL 2018; 22:217-30. [PMID: 29706061 PMCID: PMC5949124 DOI: 10.22034/ibj.22.4.217] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastric cancer (GC) is regarded as the fifth most common cancer and the third cause of cancer-related deaths worldwide. Mechanism of GC pathogenesis is still unclear and relies on multiple factors, including environmental and genetic characteristics. One of the most important environmental factors of GC occurrence is infection with Helicobacter pylori that is classified as class one carcinogens. Dysregulation of several genes and pathways play an essential role during gastric carcinogenesis. Dysregulation of developmental pathways such as Wnt/β-catenin signaling, Hedgehog signaling, Hippo pathway, Notch signaling, nuclear factor-kB, and epidermal growth factor receptor have been found in GC. Epithelial-mesenchymal transition, as an important process during embryogenesis and tumorigenesis, is supposed to play a role in initiation, invasion, metastasis, and progression of GC. Although surgery is the main therapeutic modality of the disease, the understanding of biological processes of cell signaling pathways may help to develop new therapeutic targets for GC.
Collapse
Affiliation(s)
- Fatemeh Molaei
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Yasaman Fahim
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
29
|
Kowalski-Chauvel A, Teissier G, Toulas C, Cohen-Jonathan-Moyal E, Seva C. By modulating α2β1 integrin signalling, gastrin increases adhesion oF AGS-GR gastric cancer cells. Exp Cell Res 2018; 362:498-503. [PMID: 29253536 DOI: 10.1016/j.yexcr.2017.12.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/13/2017] [Accepted: 12/14/2017] [Indexed: 01/01/2023]
Abstract
Peritoneal metastasis is a major cause of recurrence of gastric cancer and integrins are key molecules involved in gastric cancer cells attachment to the peritoneum. The peptide hormone, gastrin, initially identified for its role in gastric acid secretion is also a growth factor for gastric mucosa. Gastrin has also been shown to contribute to gastric cancers progression. Here, we provide the first evidence that gastrin increases the adhesion of gastric cancer cells. Gastrin treatment induces the expression of α2 integrin subunit through a mechanism that involves the ERK pathway. We also observed in response to gastrin an increase in the amount of α2 integrin associated with β1subunit. In addition, gastrin-stimulated cell adhesion was blocked with an anti-α2β1 integrin neutralizing antibody. We also show that gastrin activates the integrin pathway via the phosphorylation of β1 integrin by a Src family kinase. This mechanism may contribute to the enhancement of cell adhesion observed in response to gastrin since we found an inhibition of gastrin-mediated cell adhesion when cells were treated with a Src inhibitor. By regulating one of the key step of the metastatic process gastrin might contribute to increase the aggressive behaviour of human gastric tumours.
Collapse
Affiliation(s)
- Aline Kowalski-Chauvel
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, team 11, Oncopole 2 Avenue Hubert Curien, CS 53717, 31037 Toulouse, France
| | - Guy Teissier
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, team 11, Oncopole 2 Avenue Hubert Curien, CS 53717, 31037 Toulouse, France
| | - Christine Toulas
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, team 11, Oncopole 2 Avenue Hubert Curien, CS 53717, 31037 Toulouse, France; IUCT-oncopole Toulouse, France
| | - Elizabeth Cohen-Jonathan-Moyal
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, team 11, Oncopole 2 Avenue Hubert Curien, CS 53717, 31037 Toulouse, France; IUCT-oncopole Toulouse, France
| | - Catherine Seva
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, team 11, Oncopole 2 Avenue Hubert Curien, CS 53717, 31037 Toulouse, France.
| |
Collapse
|
30
|
|
31
|
Raab-Westphal S, Marshall JF, Goodman SL. Integrins as Therapeutic Targets: Successes and Cancers. Cancers (Basel) 2017; 9:E110. [PMID: 28832494 PMCID: PMC5615325 DOI: 10.3390/cancers9090110] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 12/12/2022] Open
Abstract
Integrins are transmembrane receptors that are central to the biology of many human pathologies. Classically mediating cell-extracellular matrix and cell-cell interaction, and with an emerging role as local activators of TGFβ, they influence cancer, fibrosis, thrombosis and inflammation. Their ligand binding and some regulatory sites are extracellular and sensitive to pharmacological intervention, as proven by the clinical success of seven drugs targeting them. The six drugs on the market in 2016 generated revenues of some US$3.5 billion, mainly from inhibitors of α4-series integrins. In this review we examine the current developments in integrin therapeutics, especially in cancer, and comment on the health economic implications of these developments.
Collapse
Affiliation(s)
- Sabine Raab-Westphal
- Translational In Vivo Pharmacology, Translational Innovation Platform Oncology, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany.
| | - John F Marshall
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | - Simon L Goodman
- Translational and Biomarkers Research, Translational Innovation Platform Oncology, Merck KGaA, 64293 Darmstadt, Germany.
| |
Collapse
|
32
|
Expression of Vav3 protein and its prognostic value in patients with gastric cancer. Pathol Res Pract 2017; 213:435-440. [PMID: 28285969 DOI: 10.1016/j.prp.2017.01.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/28/2017] [Accepted: 01/30/2017] [Indexed: 01/08/2023]
Abstract
Vav3 is associated with tumor growth, apoptosis, invasion, metastasis and angiogenesis. In this study, we detected the expression of Vav3 in gastric cancer tissues, and explored its role in invasion, metastasis and prognosis of gastric cancer. Vav3, MMP-2, MMP-9, TIMP-1 and TIMP-2 in primary lesion and pericarcinous tissues were tested with Immunohistochemistry and Western blot. Results showed a higher expression of Vav3 in primary lesion than in pericarcinous tissue, and the expression of Vav3 was significantly correlated with MMP-2, MMP-9 and TIMP-1 in gastric cancer tissues. Overexpression of Vav3 was associated with poorer differentiation, advanced clinical stage, more significant infiltration depth, lymphatic metastasis, and perineural invasion. Results of Kaplan-Meier verified that overexpression of Vav3 was related to poorer prognosis and shorter survival time. Moreover, Cox proportional hazard model revealed that overexpression of Vav3 was an independent risk factor of prognosis for patients with gastric cancer. In all, we conclude that overexpression of Vav3 is an independent risk factor for prognosis of gastric cancer, and can be used as a prognostic indicator. This may be because that Vav3 could regulate genes which associated with the invasion and metastasis.
Collapse
|