1
|
Lee J, Kim IH, Seol D, Lee S, Yoo M, Lee TK, Yoon SH, Lee E, Hwang D, Kang SH, Park YS, Ku B, Jeon SY, Choi Y, Jung K, Kim JW, Kim JW, Ahn SH, Lee KW, Kim HH, Oh HJ, Lee DW, Suh YS. High-Throughput Chemotherapeutic Drug Screening System for Gastric Cancer (Cure-GA). Ann Surg Oncol 2025; 32:3781-3795. [PMID: 39847282 PMCID: PMC11976768 DOI: 10.1245/s10434-024-16850-0] [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: 10/11/2024] [Accepted: 12/24/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Three dimensional (3D) cell cultures can be effectively used for drug discovery and development but there are still challenges in their general application to high-throughput screening. In this study, we developed a novel high-throughput chemotherapeutic 3D drug screening system for gastric cancer, named 'Cure-GA', to discover clinically applicable anticancer drugs and predict therapeutic responses. METHODS Primary cancer cells were isolated from 143 fresh surgical specimens by enzymatic treatment. Cell-Matrigel mixtures were automatically printed onto the micropillar surface then stabilized in an optimal culture medium for 3 days to form tumoroids. These tumoroids were exposed in the drug-containing media for 7 days. Cell viability was measured by fluorescence imaging and adenosine triphosphate assays. On average, 0.31 ± 0.23 g of fresh tumor tissue yielded 4.05×106 ± 4.38×106 viable cells per sample. RESULTS Drug response results were successfully acquired from 103 gastric cancer tissues (success rate = 72%) within 13 ± 2 days, averaging 6.4 ± 2.7 results per sample. Pearson correlation analysis showed viable cell numbers significantly impacted drug data acquisition (p < 0.00001). Tumoroids retained immunohistochemical characteristics, mutation signatures, and gene expression consistent with primary tumors. Drug reactivity data enabled prediction of synergistic drug correlations. Additionally, a multiparameter index-based prognosis model for patients undergoing gastrectomy followed by adjuvant XELOX was developed, showing significant differences in 1-year recurrence-free survival rates between drug responders and non-responders (p < 0.0001). CONCLUSIONS The Cure-GA platform enables rapid evaluation of chemotherapeutic responses using patient-derived tumoroids, providing clinicians with crucial insights for personalized treatment strategies and improving therapeutic outcomes.
Collapse
Affiliation(s)
- Jieun Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - In Hee Kim
- Department of Precision Medicine, Medical and Bio Decision (MBD Co., Ltd), Suwon, Republic of Korea
| | - Donghyeok Seol
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sangjun Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Surgery, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Mira Yoo
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - So Hee Yoon
- Department of Precision Medicine, Medical and Bio Decision (MBD Co., Ltd), Suwon, Republic of Korea
| | - Eunju Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Surgery, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, Republic of Korea
| | - Duyeong Hwang
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - So Hyun Kang
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Young Suk Park
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Bosung Ku
- Department of Precision Medicine, Medical and Bio Decision (MBD Co., Ltd), Suwon, Republic of Korea
| | - Sang Youl Jeon
- Department of Precision Medicine, Medical and Bio Decision (MBD Co., Ltd), Suwon, Republic of Korea
| | - Yongmun Choi
- Department of Precision Medicine, Medical and Bio Decision (MBD Co., Ltd), Suwon, Republic of Korea
| | - Keehoon Jung
- Department of Anatomy and Cell Biology / Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji-Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sang-Hoon Ahn
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hyung-Ho Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Surgery, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, Republic of Korea
| | - Hyeon Jeong Oh
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
| | - Dong Woo Lee
- Department of Biomedical Engineering, Gachon University, Seongnam, Republic of Korea.
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Yang L, Yi Y, Mei Z, Huang D, Tang S, Hu L, Liu L. Circular RNAs in cancer stem cells: Insights into their roles and mechanisms (Review). Int J Mol Med 2025; 55:50. [PMID: 39930823 PMCID: PMC11781527 DOI: 10.3892/ijmm.2025.5491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/03/2025] [Indexed: 02/14/2025] Open
Abstract
Cancer stem cells (CSCs) represent a small, yet pivotal subpopulation of tumor cells that play significant roles in tumor initiation, progression and therapeutic resistance. Circular RNAs (circRNAs) are a distinct class of RNAs characterized by their closed‑loop structures, lacking 5' to 3'ends. There is growing evidence that circRNAs are integral to the development and regulation of CSCs. Aberrant expression of circRNAs in CSCs can contribute to oncogenic properties and drug resistance. Specifically, oncogenic circRNAs modulate CSC behavior via key signaling pathways, thereby promoting CSC self‑renewal and maintenance, as well as tumor progression. This review summarizes the latest research on the functional roles and regulatory mechanisms of circRNAs in CSC behavior and discusses potential applications and challenges of targeting circRNAs in CSCs. Understanding the intricate interactions between circRNAs and CSCs may lead to novel therapeutic strategies that effectively combat treatment resistance and improve patient outcomes.
Collapse
Affiliation(s)
- Lunyu Yang
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Yuling Yi
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Zhu Mei
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Dongmei Huang
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Sitian Tang
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Liyi Hu
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Ling Liu
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| |
Collapse
|
3
|
Yang M, Lin W, Huang J, Mannucci A, Luo H. Novel immunotherapeutic approaches in gastric cancer. PRECISION CLINICAL MEDICINE 2024; 7:pbae020. [PMID: 39397869 PMCID: PMC11467695 DOI: 10.1093/pcmedi/pbae020] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/08/2024] [Accepted: 09/08/2024] [Indexed: 10/15/2024] Open
Abstract
Gastric cancer is a malignant tumor that ranks third in cancer-related deaths worldwide. Early-stage gastric cancer can often be effectively managed through surgical resection. However, the majority of cases are diagnosed in advanced stages, where outcomes with conventional radiotherapy and chemotherapy remain unsatisfactory. Immunotherapy offers a novel approach to treating molecularly heterogeneous gastric cancer by modifying the immunosuppressive tumor microenvironment. Immune checkpoint inhibitors and adoptive cell therapy are regarded as promising modalities in cancer immunotherapy. Food and Drug Administration-approved programmed death-receptor inhibitors, such as pembrolizumab, in combination with chemotherapy, have significantly extended overall survival in gastric cancer patients and is recommended as a first-line treatment. Despite challenges in solid tumor applications, adoptive cell therapy has demonstrated efficacy against various targets in gastric cancer treatment. Among these approaches, chimeric antigen receptor-T cell therapy research is the most widely explored and chimeric antigen receptor-T cell therapy targeting claudin18.2 has shown acceptable safety and robust anti-tumor capabilities. However, these advancements primarily remain in preclinical stages and further investigation should be made to promote their clinical application. This review summarizes the latest research on immune checkpoint inhibitors and adoptive cell therapy and their limitations, as well as the role of nanoparticles in enhancing immunotherapy.
Collapse
Affiliation(s)
- Meng Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Wuhao Lin
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jiaqian Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Alessandro Mannucci
- Gastroenterology and Gastrointestinal Emndoscopy Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan 20132, Italy
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope; Monrovia, CA 91016, USA
| | - Huiyan Luo
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| |
Collapse
|
4
|
Yu B, de Vos D, Guo X, Peng S, Xie W, Peppelenbosch MP, Fu Y, Fuhler GM. IL-6 facilitates cross-talk between epithelial cells and tumor- associated macrophages in Helicobacter pylori-linked gastric carcinogenesis. Neoplasia 2024; 50:100981. [PMID: 38422751 PMCID: PMC10912637 DOI: 10.1016/j.neo.2024.100981] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE Helicobacter pylori (H. pylori) is a significant risk factor for development of gastric cancer (GC), one of the deadliest malignancies in the world. However, the mechanism by which H. pylori induces gastric oncogenesis remains unclear. Here, we investigated the function of IL-6 in gastric oncogenesis and macrophage-epithelial cell interactions. METHODS We analyzed publicly available datasets to investigate the expression of IL-6 and infiltration of M2 macrophages in GC tissues, and determine the inter-cellular communication in the context of IL-6. Human gastric epithelial and macrophage cell lines (GES-1 and THP-1-derived macrophages, respectively) were used in mono- and co-culture experiments to investigate autocrine-and paracrine induction of IL-6 expression in response to H. pylori or IL-6 stimulation. RESULTS We found that IL-6 is highly expressed in GC and modulates survival. M2 macrophage infiltration is predominant in GC and drives an IL-6 mediated communication with gastric epithelium cells. In vitro, IL-6 triggers its own expression in GES-1 and THP-1-derived macrophages cells. In addition, these cell lines are able to upregulate each other's IL-6 levels in an autocrine fashion, which is enhanced by H. pylori stimulation. CONCLUSION This study indicates that IL-6 in the tumor microenvironment is essential for intercellular communication. We show that H. pylori enhances an IL-6-driven autocrine and paracrine positive feedback loop between macrophages and gastric epithelial cells, which may contribute to gastric carcinogenesis.
Collapse
Affiliation(s)
- Bingting Yu
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center Rotterdam, Dr Molewaterplein 40, Rotterdam, GD 3015, the Netherlands
| | - Danny de Vos
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center Rotterdam, Dr Molewaterplein 40, Rotterdam, GD 3015, the Netherlands; Department of Infectious Diseases, Leiden University Medical Centre, the Netherlands; Department of Parasitology, Leiden University Medical Centre, the Netherlands
| | - Xiaopei Guo
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center Rotterdam, Dr Molewaterplein 40, Rotterdam, GD 3015, the Netherlands
| | - SanFei Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenjie Xie
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center Rotterdam, Dr Molewaterplein 40, Rotterdam, GD 3015, the Netherlands
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center Rotterdam, Dr Molewaterplein 40, Rotterdam, GD 3015, the Netherlands.
| |
Collapse
|
5
|
Shi L, Zhang D, Han H, Zhang L, Li S, Yang F, He C. HOTAIR knockdown impairs metastasis of cervical cancer cells by down-regulating metastasis-related genes. J OBSTET GYNAECOL 2023; 43:2181060. [PMID: 36972141 DOI: 10.1080/01443615.2023.2181060] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
This study investigated the role of LncRNA HOTAIR knockdown in the biological impacts on cervical cancer cells. The HOTAIR gene in two human cervical cancer cell lines was silenced with small interfering (si) RNA siHOTAIR. Proliferation, apoptosis, migration and invasion of cells were assessed following the knockdown. The expressions of Notch1, EpCAM, E-cadherin, vimentin and STAT3 were assessed using qRT-PCR and Western blotting analysis. Compared with controls, HOTAIR levels were reduced significantly, the OD values of cells were significantly decreased in proliferation assays, cell apoptosis was significantly increased, cell migration and invasion were significantly reduced after HOTAIR knockdown. Molecular analysis showed that Notch1, EpCAM, vimentin and STAT3 expressions were decreased significantly, while the expression of E-cadherin was significantly increased after HOTAIR knockdown. Rescue experiments further confirmed that Notch1 and STAT3 were involved in siHOTAIR-mediated reduction of migration and invasion of cervical cancer cells.IMPACT STATEMENTWhat is already known on this subject? Long non-coding RNAs including HOTAIR, is implicated in occurrence and development of cancer and have been explored to develop new therapeutic options for cancer.What do the results of this study add? HOTAIR silencing significantly reduces the viability and migration ability of cells and induces cell apoptosis, adding experimental data supporting the potential use of HOTAIR specific-siRNA as a therapeutic avenue for the cancer.What are the implications of these findings for clinical practice and/or further research? The finding from this study would help develop clinically applicable therapeutic avenues for the cancer and identify new treatment targets in the relevant pathways leading to new drugs or treatments.
Collapse
Affiliation(s)
- Lei Shi
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Dehui Zhang
- Department of Oncology, Daqing Oilfield General Hospital and Huiren Cancer Hospital, Daqing, P.R. China
| | - Huijuan Han
- Department of Obstetrics and Gynecology, the 962 Hospital, Joint Logistics Support of the Chinese People's Liberation Army, Harbin, P.R. China
| | - Liangyu Zhang
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Sirui Li
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Fang Yang
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Caijun He
- Department of Obstetrics and Gynecology, the 962 Hospital, Joint Logistics Support of the Chinese People's Liberation Army, Harbin, P.R. China
| |
Collapse
|
6
|
Deng Q, Jiang B, Yan H, Wu J, Cao Z. Circulating tumor cells in gastric cancer: developments and clinical applications. Clin Exp Med 2023; 23:4385-4399. [PMID: 37548815 DOI: 10.1007/s10238-023-01158-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023]
Abstract
Circulating tumor cells (CTCs), which are shed from primary tumor or metastatic sites into the bloodstream and subsequently seed into distant tissues, are considered as the precursors of metastases. Gastric cancer (GC) is a highly heterogeneous malignant tumor. With regard to the diagnosis of GC, secondary pathological biopsy is difficult, while invasive examination is harmful to patients. In recent years, CTCs have made great progress in tumor diagnosis, prognosis prediction, efficacy detection and treatment guidance, but the research on the role of CTCs in GC remains limited. The following sections review the landmark studies demonstrating the technical approaches of CTCs monitoring in the field of GC. Moreover, we highlight the clinical application of CTCs numbers and phenotypes in monitoring the therapeutic efficacy and judging patient prognosis by sequential blood analyses.
Collapse
Affiliation(s)
- Qian Deng
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Bo Jiang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Haijiao Yan
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.
| | - Jun Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Zhenzhen Cao
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| |
Collapse
|
7
|
TROP2: a potential marker in diagnosis of thyroid neoplasms. Ir J Med Sci 2023; 192:99-103. [PMID: 35355197 DOI: 10.1007/s11845-022-02976-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVES The human trophoblast cell surface antigen (TROP-2), a transmembrane glycoprotein, has recently been investigated as a valuable marker of thyroid epithelial neoplasms. In this study, we aimed to demonstrate the diagnostic utility of TROP2 in thyroid neoplasms. METHODS A total of 308 cases, including 170 cases of different PTC variants, 50 cases of benign and non-neoplastic lesions, and 86 cases of other neoplasms (NIFT-P, poorly differentiated thyroid carcinoma, anaplastic thyroid carcinoma, follicular thyroid carcinoma, Hürthle cell carcinoma, and medullary thyroid carcinoma), were included in this study. Only membranous staining with TROP2 was considered positive. RESULTS In PTC, classical (n = 35), tall cell (n = 41), follicular (n = 39), solid (n = 19), hobnail (n = 10), Warthin-like (n = l0), columnar (n = 7), and oncocytic variant (n = 9) were 100%, 97.6%, 5.1%, 42.1%, 90%, 90%, 28.6%, and 33.3% positive, respectively. A negative reaction was observed in all 50 cases of benign and non-neoplastic lesions and also in surrounding normal thyroid tissue in all cases. Negative reactions were observed in 83 (n = 86) of other thyroid neoplasms including NIFT-P (n = 20), poorly differentiated thyroid carcinoma (n = 10), anaplastic thyroid carcinoma (n = 10), follicular thyroid carcinoma (n = 18), Hürthle cell carcinoma (n = 18), and medullary thyroid carcinoma (n = 10). Only focal positivity was observed in three Hürthle cell carcinoma cases. CONCLUSION TROP2 was a helpful marker for differentiating PTC, especially in the classic, tall cell, hobnail, and Warthin-like variants, benign and non-neoplastic lesions, and other neoplasms of the thyroid.
Collapse
|
8
|
Martin J, Islam F. Detection and Isolation of Cancer Stem Cells. CANCER STEM CELLS: BASIC CONCEPT AND THERAPEUTIC IMPLICATIONS 2023:45-69. [DOI: 10.1007/978-981-99-3185-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Mishra A, Pathak Y, Mishra SK, Prakash H, Tripathi V. Natural compounds as a potential modifier of stem cells renewal: Comparative analysis. Eur J Pharmacol 2022; 938:175412. [PMID: 36427534 DOI: 10.1016/j.ejphar.2022.175412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/09/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Cancer stem cells (CSCs) are indispensable for development, progression, drug resistance, and tumor metastasis. Current cancer-directed interventions target targeting rapidly dividing cancer cells and slow dividing CSCs, which are the root cause of cancer origin and recurrence. The most promising targets include several self-renewal pathways involved in the maintenance and renewal of CSCs, such as the Wnt/β-Catenin, Sonic Hedgehog, Notch, Hippo, Autophagy, and Ferroptosis. In view of safety, natural compounds are coming to the front line of treatment modalities for modifying various signaling pathways simultaneously involved in maintaining CSCs. Therefore, targeting CSCs with natural compounds is a promising approach to treating various types of cancers. In view of this, here we provide a comprehensive update on the current status of natural compounds that effectively tune key self-renewal pathways of CSCs. In addition, we highlighted surface expression markers in several types of cancer. We also emphasize how natural compounds target these self-renewal pathways to reduce therapy resistance and cancer recurrence properties of CSCs, hence providing valuable cancer therapeutic strategies. The inclusion of nutraceuticals is believed to enhance the therapeutic efficacy of current cancer-directed interventions significantly.
Collapse
Affiliation(s)
- Amaresh Mishra
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201310, India
| | - Yamini Pathak
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201310, India
| | | | - Hridayesh Prakash
- Amity Institute of Virology and Immunology, Amity University, Uttar Pradesh, India
| | - Vishwas Tripathi
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201310, India.
| |
Collapse
|
10
|
Fu X, Zhang W, Li S, Ling N, Yang Y, Dazhi Z. Identification of alanine aminotransferase 1 interaction network via iTRAQ-based proteomics in alternating migration, invasion, proliferation and apoptosis of HepG2 cells. Aging (Albany NY) 2022; 14:7137-7155. [PMID: 36107005 PMCID: PMC9512495 DOI: 10.18632/aging.204286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/31/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the mechanism of alanine aminotransferase 1 (ALT1) in the progression of HCC, the differentially expressed proteins (DEPs) in the ALT1 interaction network were identified by targeted proteomic analysis. METHODS Wound healing and transwell assays were conducted to assess the effect of ALT1 on cellular migration and invasion. Cell Counting Kit-8 (CCK-8), colony formation, and flow cytometry assays were performed to identify alterations in proliferation and apoptosis. After coimmunoprecipitation processing, mass spectrometry with iso-baric tags for relative and absolute quantitation was utilized to explore the protein interactions in ALT1 knockdown HepG2 cells. RESULTS The results showed that ALT1 knockdown inhibits the migration, invasion, proliferation of HepG2 cells, and promotes apoptosis. A total of 116 DEPs were identified and the bioinformatics analysis suggested that the ALT1-interacting proteins were primarily associated with cellular and metabolic processes. Knockdown of ALT1 in HepG2 cells reduced the expression of Ki67 and epithelial cell adhesion molecule (EP-CAM), while the expression of apoptosis-stimulating protein 2 of p53 (ASPP2) was increased significantly. Suppression of the ALT1 and EP-CAM expression contributed to alterations in epithelial-mesenchymal transition (EMT) -associated markers and matrix metalloproteinases (MMPs). Additionally, inhibition of ALT1 and Ki67 also decreased the expression of apoptosis and proliferation factors. Furthermore, inhibition of ALT1 and ASPP2 also changed the expression of P53, which may be the signaling pathway by which ALT regulates these biological behaviors. CONCLUSIONS This study indicated that the ALT1 protein interaction network is associated with the biological behaviors of HepG2 cells via the p53 signaling pathway.
Collapse
Affiliation(s)
- Xiao Fu
- Department of Infectious Diseases, Institute for Viral Hepatitis, Key Laboratory of Molecular Biology for Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Wenyue Zhang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong 999077, P.R. China
| | - Shiying Li
- Department of Infectious Diseases, Institute for Viral Hepatitis, Key Laboratory of Molecular Biology for Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Ning Ling
- Department of Infectious Diseases, Institute for Viral Hepatitis, Key Laboratory of Molecular Biology for Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yixuan Yang
- Department of Infectious Diseases, Institute for Viral Hepatitis, Key Laboratory of Molecular Biology for Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Zhang Dazhi
- Department of Infectious Diseases, Institute for Viral Hepatitis, Key Laboratory of Molecular Biology for Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
11
|
Huang B, Miao L, Liu J, Zhang J, Li Y. A promising antitumor method: Targeting CSC with immune cells modified with CAR. Front Immunol 2022; 13:937327. [PMID: 36032145 PMCID: PMC9403009 DOI: 10.3389/fimmu.2022.937327] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022] Open
Abstract
Tumors pose a great threat to human health; as a subgroup of tumor cells, cancer stem cells (CSCs) contribute to the genesis, development, metastasis, and recurrence of tumors because of their enhanced proliferation and multidirectional differentiation. Thus, a critical step in tumor treatment is to inhibit CSCs. Researchers have proposed many methods to inhibit or reduce CSCs, including monoclonal antibodies targeting specific surface molecules of CSCs, signal pathway inhibitors, and energy metabolic enzyme inhibitors and inducing differentiation therapy. Additionally, immunotherapy with immune cells engineered with a chimeric antigen receptor (CAR) showed favorable results. However, there are few comprehensive reviews in this area. In this review, we summarize the recent CSC targets used for CSC inhibition and the different immune effector cells (T cells, natural killer (NK) cells, and macrophages) which are engineered with CAR used for CSC therapy. Finally, we list the main challenges and options in targeting CSC with CAR-based immunotherapy. The design targeting two tumor antigens (one CSC antigen and one mature common tumor antigen) should be more reasonable and practical; meanwhile, we highlight the potential of CAR-NK in tumor treatment.
Collapse
Affiliation(s)
- Binjie Huang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Lele Miao
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Jie Liu
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Jiaxing Zhang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Yumin Li,
| |
Collapse
|
12
|
Faghfuri E, Shadbad MA, Faghfouri AH, Soozangar N. Cellular immunotherapy in gastric cancer: adoptive cell therapy and dendritic cell-based vaccination. Immunotherapy 2022; 14:475-488. [PMID: 35232264 DOI: 10.2217/imt-2021-0285] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of the most frequently diagnosed malignancies. Recent studies have highlighted cellular immunotherapy (CI) as a promising approach for treating this disease. Among the CI-based approaches, adoptive cell therapy and dendritic cell-based vaccination are commonly studied in preclinical and clinical trials. Here we review the current evidence on the potentiality of CI in treating GC, the targets for adoptive cell therapy, ongoing clinical trials, constraints and the future outlook. The results suggest that there is a need to identify novel biomarkers that predict which GC patients will most likely respond to these approaches. Also, CI plus chemotherapy or immune checkpoint inhibitors can improve the survival of patients with late-stage GC. Therefore, this approach can be promising for treating these patients.
Collapse
Affiliation(s)
- Elnaz Faghfuri
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | | | - Narges Soozangar
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
13
|
Yang Y, Meng WJ, Wang ZQ. Cancer Stem Cells and the Tumor Microenvironment in Gastric Cancer. Front Oncol 2022; 11:803974. [PMID: 35047411 PMCID: PMC8761735 DOI: 10.3389/fonc.2021.803974] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/08/2021] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) remains one of the leading causes of cancer-related death worldwide. Cancer stem cells (CSCs) might be responsible for tumor initiation, relapse, metastasis and treatment resistance of GC. The tumor microenvironment (TME) comprises tumor cells, immune cells, stromal cells and other extracellular components, which plays a pivotal role in tumor progression and therapy resistance. The properties of CSCs are regulated by cells and extracellular matrix components of the TME in some unique manners. This review will summarize current literature regarding the effects of CSCs and TME on the progression and therapy resistance of GC, while emphasizing the potential for developing successful anti-tumor therapy based on targeting the TME and CSCs.
Collapse
Affiliation(s)
| | - Wen-Jian Meng
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | | |
Collapse
|
14
|
Chang Y, Roy S, Pan Z. Store-Operated Calcium Channels as Drug Target in Gastroesophageal Cancers. Front Pharmacol 2021; 12:668730. [PMID: 34012400 PMCID: PMC8126661 DOI: 10.3389/fphar.2021.668730] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/12/2021] [Indexed: 12/24/2022] Open
Abstract
Gastroesophageal cancers, including tumors occurring in esophagus and stomach, usually have poor prognosis and lack effective chemotherapeutic drugs for treatment. The association between dysregulated store-operated calcium entry (SOCE), a key intracellular Ca2+ signaling pathway and gastroesophageal cancers are emerging. This review summarizes the recent advances in understanding the contribution of SOCE-mediated intracellular Ca2+ signaling to gastroesophageal cancers. It assesses the pathophysiological role of each component in SOCE machinery, such as Orais and STIMs in the cancer cell proliferation, migration, and invasion as well as stemness maintenance. Lastly, it discusses efforts towards development of more specific and potent SOCE inhibitors, which may be a new set of chemotherapeutic drugs appearing at the horizon, to provide either targeted therapy or adjuvant treatment to overcome drug resistance for gastroesophageal cancers.
Collapse
Affiliation(s)
- Yan Chang
- College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, United States
| | - Souvik Roy
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, United States
| | - Zui Pan
- College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, United States
| |
Collapse
|
15
|
Dinneen K, Baird AM, Ryan C, Sheils O. The Role of Cancer Stem Cells in Drug Resistance in Gastroesophageal Junction Adenocarcinoma. Front Mol Biosci 2021; 8:600373. [PMID: 33628765 PMCID: PMC7897661 DOI: 10.3389/fmolb.2021.600373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/06/2021] [Indexed: 12/24/2022] Open
Abstract
Gastroesophageal junction adenocarcinomas (GEJA) have dramatically increased in incidence in the western world since the mid-20th century. Their prognosis is poor, and conventional anti-cancer therapies do not significantly improve survival outcomes. These tumours are comprised of a heterogenous population of both cancer stem cells (CSC) and non-CSCs, with the former playing a crucial role in tumorigenesis, metastasis and importantly drug resistance. Due to the ability of CSCs to self-replicate indefinitely, their resistance to anti-cancer therapies poses a significant barrier to effective treatment of GEJA. Ongoing drug development programmes aim to target and eradicate CSCs, however their characterisation and thus identification is difficult. CSC regulation is complex, involving an array of signalling pathways, which are in turn influenced by a number of entities including epithelial mesenchymal transition (EMT), microRNAs (miRNAs), the tumour microenvironment and epigenetic modifications. Identification of CSCs commonly relies on the expression of specific cell surface markers, yet these markers vary between different malignancies and indeed are often co-expressed in non-neoplastic tissues. Development of targeted drug therapies against CSCs thus requires an understanding of disease-specific CSC markers and regulatory mechanisms. This review details the current knowledge regarding CSCs in GEJA, with particular emphasis on their role in drug resistance.
Collapse
Affiliation(s)
- Kate Dinneen
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.,Department of Histopathology, St. James's Hospital, Dublin, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Ciara Ryan
- Department of Histopathology, St. James's Hospital, Dublin, Ireland
| | - Orla Sheils
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
16
|
Wong-Arteta J, Rey M, Aragón L, Gil-Rodríguez E, Bujanda L. The utility of flow cytometry in the diagnostic work up of malignant effusions due to nonhematopoietic neoplasms. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2020; 98:504-515. [PMID: 32506689 DOI: 10.1002/cyto.b.21886] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/19/2022]
Abstract
Malignant pleural effusion and peritoneal carcinomatosis are frequent causes of effusion. Cytological evaluation (PAP-stained slides followed by immunocytochemistry, IHC, if applicable) is currently the gold standard for the diagnosis of malignant effusions, but its sensitivity varies between 40 and 80%, being a time-consuming technique. Although flow cytometry (FC) is not routinely used in the diagnosis or follow-up of nonhematopoietic neoplasms, it has the advantage of being rapidly applicable to fresh samples, potentially decreasing the time for the diagnosis. The main objective of this study was to assess the utility of FC as a confirmatory tool in the diagnosis of neoplastic effusions, based on the expression of EpCAM antibody in tumor cells versus the cytological evaluation. In this work 1,535 serous fluids were collected, of which 101 (68 pleural, 33 ascites) were selected through a screening algorithm and sent to the FC and cytological evaluation. Seventy-three of these samples (46 pleural, 27 ascites) were considered malignant as determined by clinical, cytological and radiological criteria. According to our data, 75% (55/73) of these samples were positive by Cytology/IHC and 74% (54/73) by FC. We noticed that, although the sensitivity, specificity, and area under the curve were similar, the turn-around time was shorter when using FC. Moreover, these results clearly improved by combining both techniques. We conclude that FC provides information about malignant effusions faster than immunohistochemical staining, and we believe that performing both techniques in parallel would improve diagnostic performance.
Collapse
Affiliation(s)
- Jhonatan Wong-Arteta
- Biochemistry, Donostia University Hospital, San Sebastián, País Vasco, Spain
- School of Medicine, University of the Basque Country (UPV-EHU), San Sebastián, País Vasco, Spain
- Hematology, Asuncion Clinic, Tolosa, País Vasco, Spain
| | - Mercedes Rey
- Immunology, Donostia University Hospital, San Sebastián, País Vasco, Spain
| | - Larraitz Aragón
- Immunology, Donostia University Hospital, San Sebastián, País Vasco, Spain
| | - Eva Gil-Rodríguez
- Biochemistry, Donostia University Hospital, San Sebastián, País Vasco, Spain
| | - Luis Bujanda
- School of Medicine, University of the Basque Country (UPV-EHU), San Sebastián, País Vasco, Spain
- Gastroenterology, Donostia University Hospital, San Sebastián, País Vasco, Spain
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, San Sebastián, País Vasco, Spain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| |
Collapse
|
17
|
CAR-T Cell Therapy-An Overview of Targets in Gastric Cancer. J Clin Med 2020; 9:jcm9061894. [PMID: 32560392 PMCID: PMC7355670 DOI: 10.3390/jcm9061894] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is one of the most commonly diagnosed malignancies and, unfortunately, still has a high mortality rate. Recent research points to CAR-T immunotherapy as a promising treatment for this disease. Using genetically engineered T cells designed to target a previously selected antigen, researchers are able to harness the natural anti-tumor activity of T cells. For therapy to be successful, however, it is essential to choose antigens that are present on tumor cells but not on healthy cells. In this review, we present an overview of the most important targets for CAR-T therapy in the context of GC, including their biologic function and therapeutic application. A number of clinical studies point to the following as important markers in GC: human epidermal growth factor receptor 2, carcinoembryonic antigen, mucin 1, epithelial cell adhesion molecule, claudin 18.2, mesothelin, natural-killer receptor group 2 member D, and folate receptor 1. Although these markers have been met with some success, the search for new and improved targets continues. Key among these novel biomarkers are the B7H6 ligand, actin-related protein 2/3 (ARP 2/3), neuropilin-1 (NRP-1), desmocollin 2 (DSC2), anion exchanger 1 (AF1), and cancer-related antigens CA-72-4 and CA-19-9.
Collapse
|
18
|
Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, Zhang G, Wang X, Dong Z, Chen F, Cui H. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther 2020; 5:8. [PMID: 32296030 PMCID: PMC7005297 DOI: 10.1038/s41392-020-0110-5] [Citation(s) in RCA: 1178] [Impact Index Per Article: 235.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/15/2019] [Accepted: 12/19/2019] [Indexed: 12/18/2022] Open
Abstract
Since cancer stem cells (CSCs) were first identified in leukemia in 1994, they have been considered promising therapeutic targets for cancer therapy. These cells have self-renewal capacity and differentiation potential and contribute to multiple tumor malignancies, such as recurrence, metastasis, heterogeneity, multidrug resistance, and radiation resistance. The biological activities of CSCs are regulated by several pluripotent transcription factors, such as OCT4, Sox2, Nanog, KLF4, and MYC. In addition, many intracellular signaling pathways, such as Wnt, NF-κB (nuclear factor-κB), Notch, Hedgehog, JAK-STAT (Janus kinase/signal transducers and activators of transcription), PI3K/AKT/mTOR (phosphoinositide 3-kinase/AKT/mammalian target of rapamycin), TGF (transforming growth factor)/SMAD, and PPAR (peroxisome proliferator-activated receptor), as well as extracellular factors, such as vascular niches, hypoxia, tumor-associated macrophages, cancer-associated fibroblasts, cancer-associated mesenchymal stem cells, extracellular matrix, and exosomes, have been shown to be very important regulators of CSCs. Molecules, vaccines, antibodies, and CAR-T (chimeric antigen receptor T cell) cells have been developed to specifically target CSCs, and some of these factors are already undergoing clinical trials. This review summarizes the characterization and identification of CSCs, depicts major factors and pathways that regulate CSC development, and discusses potential targeted therapy for CSCs.
Collapse
Affiliation(s)
- Liqun Yang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Pengfei Shi
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Gaichao Zhao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Jie Xu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Wen Peng
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Jiayi Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Guanghui Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Xiaowen Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Zhen Dong
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Fei Chen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, 48201, USA
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China.
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China.
| |
Collapse
|
19
|
Yang J, Isaji T, Zhang G, Qi F, Duan C, Fukuda T, Gu J. EpCAM associates with integrin and regulates cell adhesion in cancer cells. Biochem Biophys Res Commun 2020; 522:903-909. [DOI: 10.1016/j.bbrc.2019.11.152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022]
|
20
|
Wu D, Lv J, Zhao R, Wu Z, Zheng D, Shi J, Lin S, Wang S, Wu Q, Long Y, Li P, Yao Y. PSCA is a target of chimeric antigen receptor T cells in gastric cancer. Biomark Res 2020; 8:3. [PMID: 32010446 PMCID: PMC6988264 DOI: 10.1186/s40364-020-0183-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/02/2020] [Indexed: 12/23/2022] Open
Abstract
Background Gastric cancer is a deadly malignancy and is a prognostically unfavorable entity with restricted therapeutic strategies available. Prostate stem cell antigen (PSCA) is a glycosylphosphatidylinositol (GPI)-anchored cell surface protein widely expressed in bladder, prostate, and pancreatic cancers. Existing studies have thoroughly recognized the availability of utilizing anti-PSCA CAR-T cells in the treatment of metastatic prostate cancer and non-small-cell lung cancer. However, no previous study has investigated the feasibility of using anti-PSCA CAR-T cells to treat gastric cancer, irrespective of the proven expression of PSCA on the gastric cancer cell surface. Methods We determined the expression of PSCA in several primary tumor tissues and constructed third-generation anti-PSCA CAR-T cells. We then incubated anti-PSCA CAR-T cells and GFP-T cells with target tumor cell lines at E:T ratios of 2:1, 1:1, 1:2, and 1:4 to evaluate the therapeutic efficacy of anti-PSCA CAR-T cells in vitro. We also assayed canonical T cell activation markers after coculturing anti-PSCA CAR-T cells with target cell lines by flow cytometry. The detection of a functional cytokine profile was carried out via enzyme-linked immunosorbent assays. We then evaluated the antitumor activity of anti-PSCA CAR-T cells in vivo by establishing two different xenograft GC mouse models. Results Anti-PSCA CAR-T cells exhibited upregulated activation markers and increased cytokine production profiles related to T cell cytotoxicity in an antigen-dependent manner. Moreover, anti-PSCA CAR-T cells exhibited robust anti-tumor cytotoxicity in vitro. Importantly, we demonstrated that anti-PSCA CAR-T cells delivered by peritumoral injection successfully stunted tumor progression in vivo. However, intravenous administration of anti-PSCA CAR-T cells failed to reveal any therapeutic improvements. Conclusions Our findings corroborated the feasibility of anti-PSCA CAR-T cells and their efficacy against gastric cancer, implicating the potential of applying anti-PSCA CAR-T cells to treat GC patients in the clinic.
Collapse
Affiliation(s)
- Di Wu
- 1School of Life Sciences, University of Science and Technology of China, Hefei, 230027 China.,2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Jiang Lv
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,4University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049 China
| | - Ruocong Zhao
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,5Institute of Hematology, Medical College, Jinan University, Guangzhou, 510632 China
| | - Zhiping Wu
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,4University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049 China
| | - Diwei Zheng
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,4University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049 China
| | - Jingxuan Shi
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,4University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049 China
| | - Simiao Lin
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Suna Wang
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Qiting Wu
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Youguo Long
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Peng Li
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,6Hefei Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Yao Yao
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| |
Collapse
|
21
|
Epithelial Cell Adhesion Molecule in Primary Sjögren's Syndrome Patients: Characterization and Evaluation of a Potential Biomarker. J Immunol Res 2019; 2019:3269475. [PMID: 31886299 PMCID: PMC6915146 DOI: 10.1155/2019/3269475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/09/2019] [Indexed: 11/18/2022] Open
Abstract
Objective To determine the subcellular localization of epithelial cell adhesion molecule (EpCAM) in labial salivary gland (LSG) and evaluate the diagnostic use of the extracellular domain of EpCAM (EpEX) and intracellular domain (EpICD) for primary Sjögren's syndrome (pSS). Methods Immunohistochemical (IHC) analysis was conducted using EpEX and EpICD domain-specific antibodies on labial salivary gland biopsy (LSGB) from participants. Chi-square or Fisher's exact analysis, Mann-Whitney U-test, and Kruskal-Wallis test compared differences among groups. Independent risk factors of pSS were determined by multiple logistic regression analysis. Receiver-operator characteristic curves (ROC) were carried out to estimate the diagnostic value. Results Compared to non-SS controls, loss of membranous EpEX and EpICD expression was observed in LSGB of pSS patients, which occurred in parallel with increased accumulation of cytoplastic and nuclear EpICD. The subcellular EpEX/EpICD expressions were associated with various features of pSS patients, especially histopathological grade of LSGB. Furthermore, high IHC scores of membranous EpEX were independent risk factors for pSS, even for the pSS patients at early stage. The IHC scores of subcellular EpEX/EpICD were of great diagnostic value for pSS with high sensitivity (70-80%) and specificity (85-95%). Conclusion This study first found the aberrant expression pattern of EpCAM in LSG of pSS patients. The IHC scores of subcellular EpEX/EpICD were demonstrated to have the potential to act as diagnostic biomarkers for pSS.
Collapse
|
22
|
Alquicira-Hernandez J, Sathe A, Ji HP, Nguyen Q, Powell JE. scPred: accurate supervised method for cell-type classification from single-cell RNA-seq data. Genome Biol 2019; 20:264. [PMID: 31829268 PMCID: PMC6907144 DOI: 10.1186/s13059-019-1862-5] [Citation(s) in RCA: 248] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/16/2019] [Indexed: 12/18/2022] Open
Abstract
Single-cell RNA sequencing has enabled the characterization of highly specific cell types in many tissues, as well as both primary and stem cell-derived cell lines. An important facet of these studies is the ability to identify the transcriptional signatures that define a cell type or state. In theory, this information can be used to classify an individual cell based on its transcriptional profile. Here, we present scPred, a new generalizable method that is able to provide highly accurate classification of single cells, using a combination of unbiased feature selection from a reduced-dimension space, and machine-learning probability-based prediction method. We apply scPred to scRNA-seq data from pancreatic tissue, mononuclear cells, colorectal tumor biopsies, and circulating dendritic cells and show that scPred is able to classify individual cells with high accuracy. The generalized method is available at https://github.com/powellgenomicslab/scPred/.
Collapse
Affiliation(s)
- Jose Alquicira-Hernandez
- Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia.
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia.
| | - Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Genome Technology Center, Stanford University, Palo Alto, USA
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Genome Technology Center, Stanford University, Palo Alto, USA
| | - Quan Nguyen
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Joseph E Powell
- Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia.
- Faculty of Medicine, University of New South Wales, Darlinghurst, Sydney, Australia.
| |
Collapse
|
23
|
Ma X, Kang X, He L, Zhou J, Zhou J, Sturm MB, Beer DG, Kuick R, Nancarrow DJ, Appelman HD, Pang Z, Li W, Zhang C, Zhang W, Zhang Y, Wang TD, Li M. Identification of Tumor Specific Peptide as EpCAM Ligand and Its Potential Diagnostic and Therapeutic Clinical Application. Mol Pharm 2019; 16:2199-2213. [PMID: 30974945 DOI: 10.1021/acs.molpharmaceut.9b00185] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumor targeting agents are being developed for early tumor detection and therapeutics. We previously identified the peptide SNFYMPL (SNF*) and demonstrated its specific binding to human esophageal specimens of high-grade dysplasia (HGD) and adenocarcinoma with imaging ex vivo. Here, we aim to identify the target for this peptide and investigate its potential applications in imaging and drug delivery. With SNF* conjugated affinity chromatography, mass spectrum, Western blot, enzyme-linked immunosorbent assay (ELISA), and molecular docking, we found that the epithelial cell adhesion molecule (EpCAM) was the potential target of SNF*. Next, we showed that FITC-labeled SNF* (SNF*-FITC) colocalized with EpCAM antibody on the surface of esophageal adenocarcinoma cells OE33, and SNF*-FITC binding patterns significantly changed after EpCAM knockdown or exogenous EpCAM transfection. With the data from TCGA, we demonstrated that EpCAM was overexpressed in 17 types of cancers. Using colon and gastric adenocarcinoma cells and tissues as examples, we found that SNF*-FITC bound in a pattern was colocalized with EpCAM antibody, and the SNF* binding did not upregulate the EpCAM downstream Wnt signals. Subsequently, we conjugated SNF* with our previously constructed poly(histidine)-PEG/DSPE copolymer micelles. SNF* labeling significantly improved the micelle binding with colon and gastric adenocarcinoma cells in vitro, and enhanced the antitumor effects and decreased the toxicities of the micelles in vivo. In conclusion, we identified and validated SNF* as a specific peptide for EpCAM. The future potential use of SNF* peptide in multiple tumor surveillance and tumor-targeted therapeutics was demonstrated.
Collapse
Affiliation(s)
- Xiaowen Ma
- Department of Pharmacy , The 960 Hospital of Chinese People's Liberation Army , Jinan 250031 , China
| | | | | | | | | | | | | | | | - Derek J Nancarrow
- Section of Thoracic Surgery, Department of Surgery , University of Michigan Medical School , Ann Arbor , Michigan 48109 , United States
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Pathak SJ, Mueller JL, Okamoto K, Das B, Hertecant J, Greenhalgh L, Cole T, Pinsk V, Yerushalmi B, Gurkan OE, Yourshaw M, Hernandez E, Oesterreicher S, Naik S, Sanderson IR, Axelsson I, Agardh D, Boland CR, Martin MG, Putnam CD, Sivagnanam M. EPCAM mutation update: Variants associated with congenital tufting enteropathy and Lynch syndrome. Hum Mutat 2019; 40:142-161. [PMID: 30461124 PMCID: PMC6328345 DOI: 10.1002/humu.23688] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/24/2018] [Accepted: 11/14/2018] [Indexed: 12/30/2022]
Abstract
The epithelial cell adhesion molecule gene (EPCAM, previously known as TACSTD1 or TROP1) encodes a membrane-bound protein that is localized to the basolateral membrane of epithelial cells and is overexpressed in some tumors. Biallelic mutations in EPCAM cause congenital tufting enteropathy (CTE), which is a rare chronic diarrheal disorder presenting in infancy. Monoallelic deletions of the 3' end of EPCAM that silence the downstream gene, MSH2, cause a form of Lynch syndrome, which is a cancer predisposition syndrome associated with loss of DNA mismatch repair. Here, we report 13 novel EPCAM mutations from 17 CTE patients from two separate centers, review EPCAM mutations associated with CTE and Lynch syndrome, and structurally model pathogenic missense mutations. Statistical analyses indicate that the c.499dupC (previously reported as c.498insC) frameshift mutation was associated with more severe treatment regimens and greater mortality in CTE, whereas the c.556-14A>G and c.491+1G>A splice site mutations were not correlated with treatments or outcomes significantly different than random simulation. These findings suggest that genotype-phenotype correlations may be useful in contributing to management decisions of CTE patients. Depending on the type and nature of EPCAM mutation, one of two unrelated diseases may occur, CTE or Lynch syndrome.
Collapse
Affiliation(s)
- Sagar J. Pathak
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
- Rady Children's HospitalSan DiegoCalifornia
| | - James L. Mueller
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
| | - Kevin Okamoto
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
| | - Barun Das
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
| | - Jozef Hertecant
- Genetics/Metabolics ServiceTawam HospitalAl AinUnited Arab Emirates
| | | | - Trevor Cole
- West Midlands Regional Genetics Service and Birmingham Health PartnersBirmingham Women's HospitalBirminghamUK
| | - Vered Pinsk
- Division of Pediatrics, Pediatric Gastroenterology UnitSoroka University Medical Center and Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Baruch Yerushalmi
- Division of Pediatrics, Pediatric Gastroenterology UnitSoroka University Medical Center and Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Odul E. Gurkan
- Department of PediatricsGazi University School of MedicineAnkaraTurkey
| | - Michael Yourshaw
- Department of Human GeneticsUniversity of California, Los AngelesLos AngelesCalifornia
| | - Erick Hernandez
- Pediatric GastroenterologyMiami Children's Health SystemMiamiFlorida
| | | | - Sandhia Naik
- Paediatric GastroenterologyBarts and the London School of MedicineLondonUK
| | - Ian R. Sanderson
- Paediatric GastroenterologyBarts and the London School of MedicineLondonUK
| | - Irene Axelsson
- Department of PediatricsSkane University HospitalMalmoSweden
| | - Daniel Agardh
- Department of Clinical SciencesLund University, Skane University HospitalMalmoSweden
| | - C. Richard Boland
- Department of MedicineUniversity of California, San DiegoLa JollaCalifornia
| | - Martin G. Martin
- Department of PediatricsUniversity of California, Los AngelesLos AngelesCalifornia
| | - Christopher D. Putnam
- Department of MedicineUniversity of California, San DiegoLa JollaCalifornia
- San Diego BranchLudwig Institute for Cancer ResearchLa JollaCalifornia
| | - Mamata Sivagnanam
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
- Rady Children's HospitalSan DiegoCalifornia
| |
Collapse
|
25
|
Abstract
Objectives: Human trophoblast cell-surface marker (TROP-2) has been reported to be overexpressed in various human carcinomas (CAs) and suggested to be a prognostic marker for some CAs. The diagnostic utility of TROP-2 in CAs has not been explored. Methods: Immunohistochemical evaluation of TROP-2 expression on tissue microarray sections of 136 thyroid neoplasms, surgical specimens of 61 atypical thyroid follicular-patterned lesions [including 33 papillary thyroid carcinomas (PTCs), 17 atypical follicular neoplasms (AFNs), and 11 adenomatoid nodules with focal nuclear atypia (ANFNA)], and 20 benign thyroid lesions, as well as 10 cytology specimens of PTCs was performed. For comparison, immunoassay for Hector Battifora mesothelial-1 (HBME-1), galectin-3, and cytokeratin 19 was performed on the 61 atypical thyroid follicular-patterned lesions. Results: Strong membranous staining with TROP-2 was seen in 94% (33/35) of classic PTCs and 81% (30/37) of confirmed follicular variant PTCs on tissue microarray and routine surgical sections, as well as 100% (10/10) of PTCs on cytology specimens; it was not observed in follicular adenomas (n=51) or CAs (n=37), AFNs or ANFNA (n=28), benign (n=20) or normal (n=15) thyroid tissue. In contrast, the expression of HBME-1 and galectin-3 was identified in 100% (33/33) of surgical cases of PTCs and in 57% (16/28) and 50% (14/28) of AFNs and ANFNA, respectively. Conclusions: Our findings demonstrate that a membranous TROP-2 staining pattern is highly specific for PTC, which may serve as a potential diagnostic marker aiding in the accurate classification of morphologically equivocal thyroid follicular-patterned lesions.
Collapse
|
26
|
Tretter JY, Schorpp K, Luxenburger E, Trambauer J, Steiner H, Hadian K, Gires O, Niessing D. A high-content screen for small-molecule regulators of epithelial cell-adhesion molecule (EpCAM) cleavage yields a robust inhibitor. J Biol Chem 2018; 293:8994-9005. [PMID: 29700109 DOI: 10.1074/jbc.ra118.002776] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/20/2018] [Indexed: 12/30/2022] Open
Abstract
Epithelial cell-adhesion molecule (EpCAM) is a transmembrane protein that regulates cell cycle progression and differentiation and is overexpressed in many carcinomas. The EpCAM-induced mitogenic cascade is activated via regulated intramembrane proteolysis (RIP) of EpCAM by ADAM and γ-secretases, generating the signaling-active intracellular domain EpICD. Because of its expression pattern and molecular function, EpCAM is a valuable target in prognostic and therapeutic approaches for various carcinomas. So far, several immunotherapeutic strategies have targeted the extracellular domain of EpCAM. However, targeting the intracellular signaling cascade of EpCAM holds promise for specifically interfering with EpCAM's proliferation-stimulating signaling cascade. Here, using a yellow fluorescence protein-tagged version of the C-terminal fragment of EpCAM, we established a high-content screening (HCS) of a small-molecule compound library (n = 27,280) and characterized validated hits that target EpCAM signaling. In total, 128 potential inhibitors were initially identified, of which one compound with robust inhibitory effects on RIP of EpCAM was analyzed in greater detail. In summary, our study demonstrates that the development of an HCS for small-molecule inhibitors of the EpCAM signaling pathway is feasible. We propose that this approach may also be useful for identifying chemical compounds targeting other disorders involving membrane cleavage-dependent signaling pathways.
Collapse
Affiliation(s)
| | - Kenji Schorpp
- the Assay Development and Screening Platform, Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, German Center for Environmental Health, 85764 Neuherberg, Germany
| | - Elke Luxenburger
- the Department of Otorhinolaryngology, Head and Neck Surgery, Grosshadern Medical Center and
| | - Johannes Trambauer
- the Biomedical Center, Metabolic Biochemistry, Ludwig-Maximilians-University Munich, 80539 Munich, Germany
| | - Harald Steiner
- the Biomedical Center, Metabolic Biochemistry, Ludwig-Maximilians-University Munich, 80539 Munich, Germany.,the German Center for Neurodegenerative Diseases, 81377 Munich, Germany, and
| | - Kamyar Hadian
- the Assay Development and Screening Platform, Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, German Center for Environmental Health, 85764 Neuherberg, Germany
| | - Olivier Gires
- the Department of Otorhinolaryngology, Head and Neck Surgery, Grosshadern Medical Center and
| | - Dierk Niessing
- From the Institute of Structural Biology and .,the Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany
| |
Collapse
|
27
|
Chen J, Shang B, Zhang H, Zhu Z, Chen L, Wang H, Ran F, Chen Q, Chen J. Enzyme-free ultrasensitive fluorescence detection of epithelial cell adhesion molecules based on a toehold-aided DNA recycling amplification strategy. RSC Adv 2018; 8:14798-14805. [PMID: 35541343 PMCID: PMC9079946 DOI: 10.1039/c8ra01362d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/04/2018] [Indexed: 01/13/2023] Open
Abstract
Epithelial cell adhesion molecules (EpCAMs) play a significant role in tumorigenesis and tumor development. EpCAMs are considered to be tumor signaling molecules for cancer diagnosis, prognosis and therapy. Herein, an enzyme-free and highly sensitive fluorescent biosensor, with a combined aptamer-based EpCAM recognition and toehold-aided DNA recycling amplification strategy, was developed for sensitive and specific fluorescence detection of EpCAMs. Due to highly specific binding between EpCAMs and corresponding aptamers, strand a, which is released from the complex of aptamer/strand a in the presence of EpCAMs which is bound to the corresponding aptamer, triggered the toehold-mediated strand displacement process. An amplified fluorescent signal was achieved by recycling strand a for ultrasensitive EpCAM detection with a detection limit as low as 0.1 ng mL-1, which was comparable or superior to that of reported immunoassays and biosensor strategies. In addition, high selectivity towards EpCAMs was exhibited when other proteins were selected as control proteins. Finally, this strategy was successfully used for the ultrasensitive fluorescence detection of EpCAMs in human serum samples with satisfactory results. Importantly, the present strategy may be also expanded for the detection of other targets using the corresponding aptamers.
Collapse
Affiliation(s)
- Jishun Chen
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Bing Shang
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Hua Zhang
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Zhengpeng Zhu
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Long Chen
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Hongmei Wang
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Fengying Ran
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Qinhua Chen
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Jun Chen
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| |
Collapse
|
28
|
Ren H, Yang X, Yang Y, Zhang X, Zhao R, Wei R, Zhang X, Zhang Y. Upregulation of LncRNA BCYRN1 promotes tumor progression and enhances EpCAM expression in gastric carcinoma. Oncotarget 2017; 9:4851-4861. [PMID: 29435146 PMCID: PMC5797017 DOI: 10.18632/oncotarget.23585] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 12/13/2017] [Indexed: 11/25/2022] Open
Abstract
Brain cytoplasmic RNA 1 (BCYRN1), along non-coding RNA, plays a critical role in various diseases, including some cancers. However, the expression of BCYRN1 and its roles in gastric carcinoma (GC) still remain unidentified. Thus, this study employed RT-qPCR to detect expression of BCYRN1 in 85 paired GC samples and adjacent normal tissues, and performed in vitro studies to explore effects of BCYRN1 in GC cells on cell proliferation, apoptosis and migration. We found BCYRN1 was significantly upregulated in GC samples, and its expression was positively correlated with advanced TNM stage (p = 0.0012) and tumor size (p = 0.027). Functionally, BCYRN1 knockdown by siRNA could inhibit cell proliferation, induce G1/G0 cell cycle arrest, increase apoptosis and impair migratory ability of AGS cells. Moreover, the results of RT-qPCR and western blotting indicated that knockdown of BCYRN1 notably decreased the expression of epithelial cell adhesion molecules (EpCAM). Otherwise, overexpression of BCYRN1 in GC cells (BGC-823 and SGC-7901) could reverse the effects of BCYRN1 knockdown. Taken together, our data indicate for the first time that BCYRN1 acts as an oncogenic lncRNA in GC progression and may be a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Hao Ren
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, China.,Department of Laboratory, Yuhuangding Hospital, Qingdao University Medical College, Yantai, Shandong Province, China
| | - Xiaomin Yang
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yongmei Yang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Xiaoyu Zhang
- Clinical Medicine of Undergraduate, Taishan Medical University, Taian, Shandong Province, China
| | - Rui Zhao
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Ran Wei
- Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
29
|
Zhou H, Wang F, Chen H, Tan Q, Qiu S, Chen S, Jing W, Yu M, Liang C, Ye S, Tu J. Increased expression of long-noncoding RNA ZFAS1 is associated with epithelial-mesenchymal transition of gastric cancer. Aging (Albany NY) 2017; 8:2023-2038. [PMID: 27654478 PMCID: PMC5076450 DOI: 10.18632/aging.101048] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/05/2016] [Indexed: 02/07/2023]
Abstract
LncRNAs play critical roles in gastric cancer (GC). In this study, the expression of fourteen cancer related lncRNAs were investigated in paired tissues of 66 patients with GC, Realtime RT-PCR revealed that ZFAS1 was significantly upregulated. We then examined the expression of ZFAS1 in plasmas derived from 77 GC patients before- and post-operations and 60 healthy individuals, and found that circulating ZFAS1 was also upregulated in GC patients and operation can reduce its presence in plasma. To investigate the potential mechanisms, we compared the expression of ZFAS1 in multiple gastric cell lines and one normal cell line and found that ZFAS1 was up-regulated in GC cell lines. Furthermore, circulating tumor cells (CTC) were simulated by mixing GC cells with peripheral blood. After EpCAM antibody-based cell sorting, we found that the expression of ZFAS1 was positively correlated with EMT property of CTCs. In GC patient tissue samples, we found that Twist was positively correlated with ZFAS1 by immunohistochemical staining. Taken together, our results suggested that ZFAS1 was up-regulated in both tissues and plasmas of GC patients, and may be involved in regulation of EMT in GC progression. Thus, ZFAS1 might serve as a potential diagnostic marker and/or therapeutic target for GC.
Collapse
Affiliation(s)
- Hu Zhou
- Department of Clinical Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Fubing Wang
- Department of Clinical Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hao Chen
- Department of Clinical Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qian Tan
- Department of Clinical Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Shili Qiu
- Department of Clinical Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Shanshan Chen
- Department of Clinical Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wei Jing
- Department of Clinical Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Mingxia Yu
- Department of Clinical Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Chunzi Liang
- Department of Clinical Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | | | - Jiancheng Tu
- Department of Clinical Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
30
|
Dai M, Yuan F, Fu C, Shen G, Hu S, Shen G. Relationship between epithelial cell adhesion molecule (EpCAM) overexpression and gastric cancer patients: A systematic review and meta-analysis. PLoS One 2017; 12:e0175357. [PMID: 28403178 PMCID: PMC5389808 DOI: 10.1371/journal.pone.0175357] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 03/26/2017] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES The epithelial cell adhesion molecule (EpCAM) is one of the most commonly used markers of cancer stem cells (CSCs), but the clinical and prognostic significance of EpCAM in gastric cancer (GC) remains disputable. Motivated by heterogeneous and inconclusive results, we conducted a systematic review and meta-analysis to systematically summarize and elucidate the association between EpCAM overexpression and GC patients. METHODS The PubMed, Cochrane Library, Medline, Web of Knowledge and the China National Knowledge Infrastructure (CNKI) databases were searched to identify relevant studies. The RevMan 5.3 software was used for the meta-analysis. Fixed-effects or random-effects models were applied depending on the presence of heterogeneity. The pooled odds ratio (ORs) and 95% confidence intervals (CIs) were applied to estimate the associations between EpCAM and gastric cancer. For the significant heterogeneity studies, sensitivity analyses were applied based on the population to test the robustness of the pooled results and identify possible sources of heterogeneity. RESULTS A total of 11 studies including 1960 GC patients met our inclusion criteria. The results of the meta-analyses revealed that there were significant differences in EpCAM overexpression and tumour size (OR = 2.97, 95% CI: 2.13~4.13, P < 0.00001), the nature of the tissue (OR = 80.30, 95% CI: 29.21~220.81, P < 0.00001), lymph node metastasis (OR = 2.78, 95% CI: 1.23~6.27, P = 0.01), and the cumulative 5-year overall survival rate (OR = 0.54, 95% CI:0.29~0.99, P = 0.05). No significant associations were identified between EpCAM overexpression and gender (OR = 0.89, 95% CI: 0.66~1.19, P = 0.43), age (OR = 1.13, 95% CI: 0.58~2.20, P = 0.73), tumour stage (OR = 2.26, 95% CI: 0.79~6.45, P = 0.13), distant metastasis (OR = 2.15, 95% CI: 0.20~22.69, P = 0.52), TNM stage (OR = 5.14, 95% CI: 0.77~34.37, P = 0.09), Lauren type (OR = 1.18, 95% CI: 0.08~16.45, P = 0.9), differentiation (OR = 1.88, 95% CI: 0.65~5.41, P = 0.24). However, due to significant heterogeneity in tumor stage, lymph node metastasis, TNM stage, differentiation and Lauren type, these results should be taken carefully. CONCLUSIONS The meta-analysis demonstrated that the expression of EpCAM in the gastric cancer group was greater than that in the control group. Moreover, EpCAM overexpression was associated with larger tumour size, lymphnode metastasis and worse prognosis in gastric cancer. Due to significant heterogeneity, the sensitivity analysis suggests that population factor may be an important source of heterogeneity, and these results should be treated with caution. EpCAM may be useful as a novel prognostic factor, and large-scale and well-designed studies are needed to validate our results in the future.
Collapse
Affiliation(s)
- Meng Dai
- Department of Geriatrics, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Tumour Immunotherapy and Nutrition Therapy, Hefei, Anhui, China
| | - Fei Yuan
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Cuiqun Fu
- Department of Geriatrics, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Tumour Immunotherapy and Nutrition Therapy, Hefei, Anhui, China
| | - Guodong Shen
- Department of Geriatrics, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Tumour Immunotherapy and Nutrition Therapy, Hefei, Anhui, China
- * E-mail: (GDS); (GS)
| | - Shilian Hu
- Department of Geriatrics, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Tumour Immunotherapy and Nutrition Therapy, Hefei, Anhui, China
| | - Gan Shen
- Department of Geriatrics, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Tumour Immunotherapy and Nutrition Therapy, Hefei, Anhui, China
- * E-mail: (GDS); (GS)
| |
Collapse
|
31
|
Abbaszadegan MR, Bagheri V, Razavi MS, Momtazi AA, Sahebkar A, Gholamin M. Isolation, identification, and characterization of cancer stem cells: A review. J Cell Physiol 2017; 232:2008-2018. [PMID: 28019667 DOI: 10.1002/jcp.25759] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 12/17/2022]
Abstract
Cancer stem cells (CSCs) or tumor-initiating cells (TICs) as a small subset of neoplastic cells are able to produce a tumor (tumorigenesis), maintain the population of tumorigenic cells (self-renewal), and generate the heterogeneous cells constructing the entire tumor (pluripotency). The research on stationary and circulating CSCs due to resistance to conventional therapies and inability in complete eradication of cancer is critical for developing novel therapeutic strategies for a more effective reduction in the risk of tumor metastasis and cancer recurrence. This review compiles information about different methods of detection and dissociation, side population, cellular markers, and establishment culture of CSCs, as well as characteristics of CSCs such as tumorigenicity, and signaling pathways associated with self-renewal and the capability of the same histological tumor regeneration in various cancers.
Collapse
Affiliation(s)
- Mohammad Reza Abbaszadegan
- Human Genetic Division, Immunology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Medical Genetics Research Center, Faculty of Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Bagheri
- Human Genetic Division, Immunology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahya Shariat Razavi
- Human Genetic Division, Immunology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Biology, Faculty of Sciences, University of Sistan and Baluchestan, Zahedan, Iran
| | - Amir Abbas Momtazi
- Student Research Committee, Nanotechnology Research Center, Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehran Gholamin
- Human Genetic Division, Immunology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Laboratory Sciences, Faculty of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
32
|
Highly tumorigenic hepatocellular carcinoma cell line with cancer stem cell-like properties. PLoS One 2017; 12:e0171215. [PMID: 28152020 PMCID: PMC5289561 DOI: 10.1371/journal.pone.0171215] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/18/2017] [Indexed: 12/21/2022] Open
Abstract
There are limited numbers of models to study hepatocellular carcinoma (HCC) in vivo in immunocompetent hosts. In an effort to develop a cell line with improved tumorigenicity, we derived a new cell line from Hepa1-6 cells through an in vivo passage in C57BL/6 mice. The resulting Dt81Hepa1-6 cell line showed enhanced tumorigenicity compared to Hepa1-6 with more frequent (28±12 vs. 0±0 lesions at 21 days) and more rapid tumor development (21 (100%) vs. 70 days (10%)) in C57BL/6 mice. The minimal Dt81Hepa1-6 cell number required to obtain visible tumors was 100,000 cells. The Dt81Hepa1-6 cell line showed high hepatotropism with subcutaneous injection leading to liver tumors without development of tumors in lungs or spleen. In vitro, Dt81Hepa1-6 cells showed increased anchorage-independent growth (34.7±6.8 vs. 12.3±3.3 colonies; P<0.05) and increased EpCAM (8.7±1.1 folds; P<0.01) and β-catenin (5.4±1.0 folds; P<0.01) expression. A significant proportion of Dt81Hepa1-6 cells expressed EpCAM compared to Hepa1-6 (34.8±1.1% vs 0.9±0.13%; P<0.001). Enriched EpCAM+ Dt81Hepa1-6 cells led to higher tumor load than EpCAM- Dt81Hepa1-6 cells (1093±74 vs 473±100 tumors; P<0.01). The in vivo selected Dt81Hepa1-6 cell line shows high liver specificity and increased tumorigenicity compared to Hepa1-6 cells. These properties are associated with increased expression of EpCAM and β-catenin confirming that EpCAM+ HCC cells comprise a subset with characteristics of tumor-initiating cells with stem/progenitor cell features. The Dt81Hepa1-6 cell line with its cancer stem cell-like properties will be a useful tool for the study of hepatocellular carcinoma in vivo.
Collapse
|
33
|
Chen XL, Chen XZ, Wang YG, He D, Lu ZH, Liu K, Zhang WH, Wang W, Li CC, Xue L, Zhao LY, Yang K, Liu JP, Zhou ZG, Hu JK, Mo XM. Clinical significance of putative markers of cancer stem cells in gastric cancer: A retrospective cohort study. Oncotarget 2016; 7:62049-62069. [PMID: 27557490 PMCID: PMC5308710 DOI: 10.18632/oncotarget.11384] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 08/08/2016] [Indexed: 02/05/2023] Open
Abstract
Cancer stem cells (CSCs) are thought as the source of tumor maintaining and many CSCs markers have been identified. Regarding the heterogeneity in gastric cancer (GC), TNM stage is not enough to accurately predict the prognosis. The aim of this study was to investigate the clinical significance of CSCs markers (Lgr5, Oct4, CD133, EpCAM, CD54 and Sox2) and establish a new model based on these markers to accurately predict prognosis of GC. We retrospectively enrolled 377 GC tissues from January 2006 to October 2012 to perform immunohistochemistry (IHC), and 93 pairs of GC tissues and corresponding adjacent normal gastric tissues to perform quantitative PCR (qPCR) from December 2011 to October 2012. The clinicopathological and follow-up characteristics were collected. In IHC, Oct4, CD133 and EpCAM were independently related to tumor progression, while Sox2 were associated with well or moderate differentiation (all p<0.05). Cox regression showed that Oct4-EpCAM was an independently prognostic factor, indicating that double low expression of Oct4-EpCAM group had significantly better prognosis than control group (p=0.035). Regarding qPCR, CD133 was an independent prognostic factor, showing that the prognosis of patients with CD133 high expression was significantly worse than that of patients with CD133 low expression (p<0.001). The prognostic prediction accuracy of nomogram based on Oct4-EpCAM expression in IHC was significantly better than TNM stage alone (p=0.003). Low expressions of Oct4-EpCAM in IHC and CD133 in qPCR were favorable prognostic factors in GC. The nomogram based on Oct4-EpCAM was valuable in prognostic prediction of GC patients.
Collapse
Affiliation(s)
- Xiao-Long Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin-Zu Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi-Gao Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Du He
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zheng-Hao Lu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kai Liu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei-Han Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chang-Chun Li
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lian Xue
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin-Yong Zhao
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kun Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jian-Ping Liu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zong-Guang Zhou
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Digestive Surgery, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jian-Kun Hu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xian-Ming Mo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
34
|
Gao Y, Gu S, Zhang Y, Xie X, Yu T, Lu Y, Zhu Y, Chen W, Zhang H, Dong H, Sinko PJ, Jia L. The Architecture and Function of Monoclonal Antibody-Functionalized Mesoporous Silica Nanoparticles Loaded with Mifepristone: Repurposing Abortifacient for Cancer Metastatic Chemoprevention. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:2595-608. [PMID: 27027489 DOI: 10.1002/smll.201600550] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 02/26/2016] [Indexed: 05/23/2023]
Abstract
The circulating tumor cells (CTCs) existing in cancer survivors are considered the root cause of cancer metastasis. To prevent the devastating metastasis cascade from initiation, we hypothesize that a biodegradable nanomaterial loaded with the abortifacient mifepristone (MIF) and conjugated with the epithelial cell adhesion molecule antibody (aEpCAM) may serve as a safe and effective cancer metastatic preventive agent by targeting CTCs and preventing their adhesion-invasion to vascular intima. It is demonstrated that MIF-loaded mesoporous silica nanoparticles (MSN) coated with aEpCAM (aE-MSN-M) can specifically target and bind colorectal cancer cells in either cell medium or blood through EpCAM recognition proven by quantitative flow cytometric detection and free aEpCAM competitive assay. The specific binding results in downregulation of the captured cells and drives them into G0/G1 phase primarily attributed to the effect of aEpCAM. The functional nanoparticles significantly inhibit the heteroadhesion between cancer cells and endothelial cells, suggesting the combined inhibition effects of aEpCAM and MIF on E-selectin and ICAM-1 expression. The functionalized nanoparticles circulate in mouse blood long enough to deliver MIF and inhibit lung metastasis. The present proof-of-concept study shows that the aE-MSN-M can prevent cancer metastasis by restraining CTC activity and their adhesion-invasion to vascular intima.
Collapse
Affiliation(s)
- Yu Gao
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350002, China
| | - Songen Gu
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350002, China
| | - Yingying Zhang
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350002, China
| | - Xiaodong Xie
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350002, China
| | - Ting Yu
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350002, China
| | - Yusheng Lu
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350002, China
| | - Yewei Zhu
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350002, China
| | - Wenge Chen
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350002, China
| | - Huijuan Zhang
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350002, China
| | - Haiyan Dong
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350002, China
| | - Patrick J Sinko
- Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854-8020, USA
| | - Lee Jia
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, 350002, China
| |
Collapse
|
35
|
Gastric cancer stem cells: evidence, potential markers, and clinical implications. J Gastroenterol 2016; 51:313-26. [PMID: 26428661 DOI: 10.1007/s00535-015-1125-5] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/13/2015] [Indexed: 02/04/2023]
Abstract
Gastric cancer is a significant global health problem. It is the fifth most common cancer and third leading cause of cancer-related death worldwide (Torre et al. in CA Cancer J Clin 65(2):87-108, 2015). Despite advances in treatment, overall prognosis remains poor, due to tumour relapse and metastasis. There is an urgent need for novel therapeutic approaches to improve clinical outcomes in gastric cancer. The cancer stem cell (CSC) model has been proposed to explain the high rate of relapse and subsequent resistance of cancer to current systemic treatments (Vermeulen et al. in Lancet Oncol 13(2):e83-e89, 2012). CSCs have been identified in many solid malignancies, including gastric cancer, and have significant clinical implications, as targeting the CSC population may be essential in preventing the recurrence and spread of a tumour (Dewi et al. in J Gastroenterol 46(10):1145-1157, 2011). This review seeks to summarise the current evidence for CSC in gastric cancer, with an emphasis on candidate CSC markers, clinical implications, and potential therapeutic approaches.
Collapse
|
36
|
Xiao YB, Xi HQ, Li JY, Chen L. Expression of epithelial cellular adhesion molecule in gastric cancer: A meta-analysis. World J Meta-Anal 2016; 4:1-9. [DOI: 10.13105/wjma.v4.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 12/01/2015] [Accepted: 01/31/2016] [Indexed: 02/05/2023] Open
Abstract
AIM: To obtain an accurate evaluation of the association between high expression of epithelial cellular adhesion molecule (EpCAM) and gastric cancer (GC) risk.
METHODS: Studies that had examined the association between high expression of EpCAM and GC risk were identified by searching electronic databases PubMed, EMBASE, Cochrane library and Chinese Biomedical Literature database. Risk ratios (RRs) together with their 95%CIs were used to assess the association between high expression of EpCAM and GC risk. We selected eligible studies based on inclusion criteria. RevMan 5.3 software was used to calculate the pooled values.
RESULTS: A total of 14 studies were included in this meta-analysis. EpCAM-positive cases were significantly associated with tumor size (RR: 1.68, 95%CI: 1.47-1.91, P < 0.00001 fixed-effect), depth of invasion (RR: 1.37, 95%CI: 1.11-1.68, P = 0.003 random-effect), TNM stage (RR: 2.02, 95%CI: 1.35-3.02, P = 0.0007 random-effect), tumor location (RR: 0.80, 95%CI: 0.71-0.91, P = 0.0007 fixed-effect), histologic differentiation (RR: 1.23, 95%CI: 1.13-1.33, P < 0.00001 fixed-effect) and lymph node metastasis (RR: 1.89, 95%CI: 1.28-2.80, P = 0.001 random-effect). However, we did not observe any significant association between the presence of EpCAM with age, gender, distant metastasis, Borrmann type or Lauren classification. Additionally, EpCAM expression was not associated with the overall survival rate. The pooled HR of the overall effect was 1.39 (95%CI: 0.30-6.48, P = 0.67 random-effect).
CONCLUSION: Our meta-analysis indicates that EpCAM contributes to GC risk, which acts as a prognostic factor and a marker of poor outcome.
Collapse
|
37
|
Caron TJ, Scott KE, Fox JG, Hagen SJ. Tight junction disruption: Helicobacter pylori and dysregulation of the gastric mucosal barrier. World J Gastroenterol 2015; 21:11411-11427. [PMID: 26523106 PMCID: PMC4616217 DOI: 10.3748/wjg.v21.i40.11411] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 06/26/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
Long-term chronic infection with Helicobacter pylori (H. pylori) is a risk factor for gastric cancer development. In the multi-step process that leads to gastric cancer, tight junction dysfunction is thought to occur and serve as a risk factor by permitting the permeation of luminal contents across an otherwise tight mucosa. Mechanisms that regulate tight junction function and structure in the normal stomach, or dysfunction in the infected stomach, however, are largely unknown. Although conventional tight junction components are expressed in gastric epithelial cells, claudins regulate paracellular permeability and are likely the target of inflammation or H. pylori itself. There are 27 different claudin molecules, each with unique properties that render the mucosa an intact barrier that is permselective in a way that is consistent with cell physiology. Understanding the architecture of tight junctions in the normal stomach and then changes that occur during infection is important but challenging, because most of the reports that catalog claudin expression in gastric cancer pathogenesis are contradictory. Furthermore, the role of H. pylori virulence factors, such as cytotoxin-associated gene A and vacoulating cytotoxin, in regulating tight junction dysfunction during infection is inconsistent in different gastric cell lines and in vivo, likely because non-gastric epithelial cell cultures were initially used to unravel the details of their effects on the stomach. Hampering further study, as well, is the relative lack of cultured cell models that have tight junction claudins that are consistent with native tissues. This summary will review the current state of knowledge about gastric tight junctions, normally and in H. pylori infection, and make predictions about the consequences of claudin reorganization during H. pylori infection.
Collapse
|
38
|
Subramanian N, Kanwar JR, Kanwar RK, Krishnakumar S. Targeting Cancer Cells Using LNA-Modified Aptamer-siRNA Chimeras. Nucleic Acid Ther 2015; 25:317-22. [PMID: 26334953 DOI: 10.1089/nat.2015.0550] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Aptamers are chimerized with drug or antisense oligos or nanoparticles to generate targeted therapeutics for cancer. Aptamer chimerized siRNA rescues nonspecific delivery and, thereby, enhances the availability of siRNA to target cells. EpCAM RNA aptamer (EpApt or Ep) has potential for siRNA chimerization due to its secondary structure. Stathmin and survivin proteins are reported to aid oncogenicity in retinoblastoma (RB), breast cancer and other cancers. Thus, chimerization of EpCAM Apt with siRNA against survivin and stathmin, respectively, was performed by incorporating Locked Nucleic Acid (LNA) modification. The LNA-modified chimeric aptamers were stable until 96 h and got internalized into RB, WERI-Rb1 and breast cancer, MDAMB453 cell lines. The constructs were studied using the recombinant dicer enzyme for the siRNA generation. Quantitative polymerase chain reaction and immunofluorescence by microscopic analysis of chimeras in vitro exhibited silencing of stathmin and survivin in the RB and breast cancer model. The chimeric constructs showed significant inhibition of cell proliferation of breast cancer cells. Thus, LNA-modified aptamer-based siRNA delivery aids in cell targeting and necessitates further studies in animal models.
Collapse
Affiliation(s)
- Nithya Subramanian
- 1 Department of Nanobiotechnology, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology , Chennai, Tamil Nadu, India .,2 Nanomedicine Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Centre of Molecular and Medical Research (C-MMR), Faculty of Health, Deakin University , Geelong, Victoria, Australia
| | - Jagat R Kanwar
- 2 Nanomedicine Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Centre of Molecular and Medical Research (C-MMR), Faculty of Health, Deakin University , Geelong, Victoria, Australia
| | - Rupinder K Kanwar
- 2 Nanomedicine Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Centre of Molecular and Medical Research (C-MMR), Faculty of Health, Deakin University , Geelong, Victoria, Australia
| | - Subramanian Krishnakumar
- 1 Department of Nanobiotechnology, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology , Chennai, Tamil Nadu, India .,3 Department of L&T Ocular Pathology, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology , Chennai, Tamil Nadu, India
| |
Collapse
|
39
|
Subramanian N, Kanwar JR, Kanwar RK, Sreemanthula J, Biswas J, Khetan V, Krishnakumar S. EpCAM Aptamer-siRNA Chimera Targets and Regress Epithelial Cancer. PLoS One 2015; 10:e0132407. [PMID: 26176230 PMCID: PMC4503753 DOI: 10.1371/journal.pone.0132407] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 06/14/2015] [Indexed: 01/03/2023] Open
Abstract
Epithelial cell adhesion molecule (EpCAM), a cancer stem cell (CSC) marker is over expressed in epithelial cancers and in retinoblastoma (RB). We fabricated an EpCAM targeting aptamer-siRNA chimera and investigated its anti-tumor property and EpCAM intracellular domain (EpICD) mediated signaling in epithelial cancer. The anti-tumor efficacy of EpCAM aptamer-siEpCAM chimera (EpApt-siEp) was evaluated by qPCR, northern and Western blotting in WERI-Rb1- RB cell line, primary RB tumor cells and in MCF7- breast cancer cell line. Anti-tumor activity of EpApt-siEp was studied in vivo using epithelial cancer (MCF7) mice xenograft model. The mechanism and pathways involved in the anti-tumor activity was further studied using protein arrays and qPCR. EpApt-siEp chimera was processed in vitro by dicer enzyme. Treatment of the WERI-Rb1 and MCF7 cells with EpApt-siEp revealed statistically significant down regulation of EpCAM expression (P<0.005) and concomitant reduction in cellular proliferation. In primary RB cells cultured from RB tumors, EpApt-siEp silenced EpCAM, significantly inhibited (P<0.01) cell proliferation and induced cytotoxicity. Knockdown of EpICD expressed in RB primary tumors led to repression of pluripotency markers, SOX2, OCT4, NANOG, and CD133. In vivo studies showed complete tumor growth regression without any toxicity in animals (P<0.001) and tumor tissues showed significant downregulation (P<0.05) of EpCAM, MRP1, ABCG2, stathmin, survivin and upregulation of ATM (P<0.05) leading to apoptosis by intrinsic pathway with minor alteration in cytokines. Our results revealed that EpApt-siEp potentially eradicated EpCAM positive cancer cells through CSC marker suppression and apoptosis, while sparing normal EpCAM negative adjacent cells.
Collapse
Affiliation(s)
- Nithya Subramanian
- Department of Nanobiotechnology, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Chennai, India
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Centre for Molecular and Medical Research (C-MMR), Faculty of Health, Deakin University, Geelong, Victoria-3217, Australia
| | - Jagat R. Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Centre for Molecular and Medical Research (C-MMR), Faculty of Health, Deakin University, Geelong, Victoria-3217, Australia
- * E-mail: (SK); (JK)
| | - Rupinder K. Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Centre for Molecular and Medical Research (C-MMR), Faculty of Health, Deakin University, Geelong, Victoria-3217, Australia
| | - JagadeeshBabu Sreemanthula
- L & T Ocular Pathology department, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Chennai, India
| | - Jyotirmay Biswas
- L & T Ocular Pathology department, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Chennai, India
| | - Vikas Khetan
- Department of Ocular Oncology and Vitreoretina, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Subramanian Krishnakumar
- Department of Nanobiotechnology, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Chennai, India
- L & T Ocular Pathology department, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Chennai, India
- * E-mail: (SK); (JK)
| |
Collapse
|
40
|
Yoshida K, Tsujimoto H, Matsumura K, Kinoshita M, Takahata R, Matsumoto Y, Hiraki S, Ono S, Seki S, Yamamoto J, Hase K. CD47 is an adverse prognostic factor and a therapeutic target in gastric cancer. Cancer Med 2015; 4:1322-33. [PMID: 26077800 PMCID: PMC4567017 DOI: 10.1002/cam4.478] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 04/22/2015] [Accepted: 04/24/2015] [Indexed: 01/24/2023] Open
Abstract
CD47 is an antiphagocytic molecule that acts via ligation to signal regulatory protein alpha on phagocytes; its enhanced expression and therapeutic targeting have recently been reported for several malignancies. However, CD47 expression in gastric cancer is not well documented. Immunohistochemical expression of CD47 in surgical specimens was investigated. Expression of CD47 and CD44, a known gastric cancer stem cell marker, were investigated in gastric cancer cell lines by flow cytometry. MKN45 and MKN74 gastric cancer cells were sorted by fluorescence-activated cell sorting according to CD44 and CD47 expression levels, and their in vitro proliferation, spheroid-forming capacity, and in vivo tumorigenicity were studied. In vitro phagocytosis of cancer cells by human macrophages in the presence of a CD47 blocking monoclonal antibody (B6H12) and the survival of immunodeficient mice intraperitoneally engrafted with MKN45 cells and B6H12 were compared to experiments using control antibodies. Immunohistochemistry of the clinical specimens indicated that CD47 was positive in 57 out of 115 cases, and its positivity was an independent adverse prognostic factor. Approximately 90% of the MKN45 and MKN74 cells expressed CD47 and CD44. CD47hi gastric cancer cells showed significantly higher proliferation and spheroid colony formation than CD47lo, and CD44hiCD47hi cells showed the highest proliferation in vitro and tumorigenicity in vivo. B6H12 significantly enhanced in vitro phagocytosis of cancer cells by human macrophages and prolonged the survival of intraperitoneal cancer dissemination in mice compared to control antibodies. In conclusion, CD47 is an adverse prognostic factor and promising therapeutic target in gastric cancer.
Collapse
Affiliation(s)
- Kazumichi Yoshida
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hironori Tsujimoto
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Kouji Matsumura
- Laboratory Center, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Manabu Kinoshita
- Department of Immunology and microbiology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Risa Takahata
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Yusuke Matsumoto
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Shuichi Hiraki
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Satoshi Ono
- Division of Traumatology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Shuhji Seki
- Department of Immunology and microbiology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Junji Yamamoto
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Kazuo Hase
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
41
|
Li Y, Zhang X, Ge S, Gao J, Gong J, Lu M, Zhang Q, Cao Y, Wang DD, Lin PP, Shen L. Clinical significance of phenotyping and karyotyping of circulating tumor cells in patients with advanced gastric cancer. Oncotarget 2015; 5:6594-602. [PMID: 25026283 PMCID: PMC4196148 DOI: 10.18632/oncotarget.2175] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Karyotyping and phenotyping of circulating tumor cells (CTCs) in therapeutic cancer patients is of particular clinical significance in terms of both identifying chemo-resistant CTC subtypes and understanding CTC evolution. METHODS The integrated subtraction enrichment (SET) and immunostaining-fluorescence in situ hybridization (iFISH) platform was applied to detect and characterize CTCs in patients with advanced gastric cancer (AGC). Status of human epidermal growth factor receptor 2 (HER2) expressing and aneuploidy of chromosome 8 in CTCs enriched from the patients was examined by SET-iFISH following clinical chemotherapy or HER2-targeted therapy. CellSearch system was applied as a reference control. RESULTS Phenotyping of CTCs in HER2 positive AGC patients demonstrated that HER2+ CTCs could be effectively eliminated in response to HER2-targeted therapy. Karyotyping of CTCs indicated that distinct CTCs with different ploidies of chromosomes 8 in AGC patients correlated to either sensitivity or resistance of paclitaxel or cisplatin-based chemotherapy. Examination of the copy number of chromosome 8 in CTCs provides a potential approach for predicting chemotherapeutic efficacy and monitoring chemo-resistance. CONCLUSIONS Phenotyping and karyotyping of the enriched CTCs upon ploidy of chromosome 8 or HER2 expression is of clinical potential for monitoring chemo-resistance and evaluating therapeutic efficacy for AGC patients.
Collapse
Affiliation(s)
- Yilin Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaotian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Sai Ge
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jing Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jifang Gong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ming Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Qiyue Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yanshuo Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | | | | | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
42
|
Abstract
Context
Accurate classification of follicular-patterned thyroid lesions is not always an easy task on routine surgical hematoxylin-eosin–stained or cytologic fine-needle aspiration specimens. The diagnostic challenges are partially due to differential diagnostic criteria that are often subtle and subjective. In the past decades, tremendous advances have been made in molecular gene profiling of tumors and diagnostic immunohistochemistry, aiding in diagnostic accuracy and proper patient management.
Objective
To evaluate the diagnostic utility of the most commonly studied immunomarkers in the field of thyroid pathology by review of the literature, using the database of indexed articles in PubMed (US National Library of Medicine) from 1976–2013.
Data Sources
Literature review, authors' research data, and personal practice experience.
Conclusions
The appropriate use of immunohistochemistry by applying a panel of immunomarkers and using a standardized technical and interpretational method may complement the morphologic assessment and aid in the accurate classification of difficult thyroid lesions.
Collapse
Affiliation(s)
- Haiyan Liu
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania
| | - Fan Lin
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania
| |
Collapse
|
43
|
A novel COX-independent mechanism of sulindac sulfide involves cleavage of epithelial cell adhesion molecule protein. Exp Cell Res 2014; 326:1-9. [DOI: 10.1016/j.yexcr.2014.05.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/12/2014] [Accepted: 05/14/2014] [Indexed: 12/30/2022]
|
44
|
Tsujiura M, Ichikawa D, Konishi H, Komatsu S, Shiozaki A, Otsuji E. Liquid biopsy of gastric cancer patients: Circulating tumor cells and cell-free nucleic acids. World J Gastroenterol 2014; 20:3265-3286. [PMID: 24696609 PMCID: PMC3964398 DOI: 10.3748/wjg.v20.i12.3265] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/27/2013] [Accepted: 02/20/2014] [Indexed: 02/06/2023] Open
Abstract
To improve the clinical outcomes of cancer patients, early detection and accurate monitoring of diseases are necessary. Numerous genetic and epigenetic alterations contribute to oncogenesis and cancer progression, and analyses of these changes have been increasingly utilized for diagnostic, prognostic and therapeutic purposes in malignant diseases including gastric cancer (GC). Surgical and/or biopsy specimens are generally used to understand the tumor-associated alterations; however, those approaches cannot always be performed because of their invasive characteristics and may fail to reflect current tumor dynamics and drug sensitivities, which may change during the therapeutic process. Therefore, the importance of developing a non-invasive biomarker with the ability to monitor real-time tumor dynamics should be emphasized. This concept, so called “liquid biopsy”, would provide an ideal therapeutic strategy for an individual cancer patient and would facilitate the development of “tailor-made” cancer management programs. In the blood of cancer patients, the presence and potent utilities of circulating tumor cells (CTCs) and cell-free nucleic acids (cfNAs) such as DNA, mRNA and microRNA have been recognized, and their clinical relevance is attracting considerable attention. In this review, we discuss recent developments in this research field as well as the relevance and future perspectives of CTCs and cfNAs in cancer patients, especially focusing on GC.
Collapse
|
45
|
Plant-derived mAbs have effective anti-cancer activities by increasing ganglioside expression in colon cancers. Biotechnol Lett 2013; 35:2031-8. [PMID: 24078119 DOI: 10.1007/s10529-013-1318-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/30/2013] [Indexed: 10/26/2022]
Abstract
An epithelial cell adhesion molecule (EpCAM) was selectively expressed in human colorectal carcinoma. Treatment with plant-derived anti-EpCAM mAb (mAbP CO17-1A) and RAW264.7 cells inhibited cell growth in the human colorectal cancer cell line SW620. In SW620 treated with mAbP CO17-1A and RAW264.7 cells, expression of p53 and p21 increased, whereas the expression of G1 phase-related proteins, cyclin D1, CDK4, cyclin E, and CDK2, decreased, similar to mammalian-derived mAb (mAbM) CO17-1A. Similar to mAbM CO17-1A, treatment with mAbP CO17-1A and RAW264.7 cell decreased the expression of anti-apoptotic protein, Bcl-2, but the expression of pro-apoptotic proteins Bax, TNF-α, caspase-3, caspase-6, caspase-8 and caspase-9, increased. Cells treated with mAbP CO17-1A and RAW264.7 cells expressed metastasis-related gangliosides, GM1 and GD1a, similar to mAbM CO17-1A. These results suggest that mAbP CO17-1A is as effective on anti-cancer activity as mAbM CO17-1A.
Collapse
|
46
|
Wang YY, Li L, Zhao ZS, Wang YX, Ye ZY, Tao HQ. L1 and epithelial cell adhesion molecules associated with gastric cancer progression and prognosis in examination of specimens from 601 patients. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2013; 32:66. [PMID: 24422715 PMCID: PMC3850006 DOI: 10.1186/1756-9966-32-66] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 09/02/2013] [Indexed: 12/31/2022]
Abstract
Background L1 cell adhesion molecule (L1CAM) and epithelial cell adhesion molecule (EPCAM) have been implicated in the development and progression of gastric cancer. The present study investigated the clinical significance of L1CAM and EPCAM in the development, progression and prognosis of gastric cancer. Methods Expression of L1CAM and EPCAM were examined immunochemically in 601 clinicopathologically characterized gastric cancer cases. Results L1CAM protein was detected in 23.9% of human non-tumor mucosa samples. All samples expressed L1CAM protein at low levels. High expression of L1CAM protein was detected in 163 (27.1%) tumors. Expression of L1CAM correlated with age, tumor location, size of tumors, Lauren’s classification, depth of invasion, lymph node and distant metastases, regional lymph node stage, Tumor-Node-Metastasis (TNM) stage and prognosis. EPCAM protein was detected in 45.7% of human non-tumor mucosa samples. All samples expressed EPCAM protein at low levels. High expression of EPCAM protein was detected in 247 (41.1%) tumors. Expression of EPCAM correlated with age, tumor location, size of tumors, Lauren’s classification, depth of invasion, lymph node and distant metastases, regional lymph node stage, TNM stage and prognosis. Cumulative 5-year survival rates of patients with high expression of both L1CAM and EPCAM were significantly lower than in patients with low expression of both. Conclusions Expression of L1CAM and EPCAM in gastric cancer was significantly associated with lymph node and distant metastasis, and poor prognosis. L1CAM and EPCAM proteins could be useful markers to predict tumor progression and prognosis.
Collapse
|
47
|
Zhang ZY, Ge HY. Micrometastasis in gastric cancer. Cancer Lett 2013; 336:34-45. [DOI: 10.1016/j.canlet.2013.04.021] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 04/16/2013] [Accepted: 04/16/2013] [Indexed: 12/19/2022]
|
48
|
Kroepil F, Dulian A, Vallböhmer D, Geddert H, Krieg A, Vay C, Topp SA, am Esch JS, Baldus SE, Gires O, Knoefel WT, Stoecklein NH. High EpCAM expression is linked to proliferation and lauren classification in gastric cancer. BMC Res Notes 2013; 6:253. [PMID: 23830302 PMCID: PMC3724596 DOI: 10.1186/1756-0500-6-253] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 06/21/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The association of EpCAM expression with the progression of gastric cancer remains unclear. Here, we investigated the expression of EpCAM in gastric cancer subtypes and correlated the data to tumor cell proliferation and clinicopathologic factors. METHODS The intratumoral expression of EpCAM was assessed in 163 primary gastric cancers (61 diffuse-, 62 intestinal-, 32 mixed-type and 8 unclassified tumors) by immunohistochemistry, using the monoclonal antibody Ber-EP4. Intensity of staining was classified according the HercepTest-score using a standardized scoring system. Ki-67 was used to examine the proliferation in tumor tissue. RESULTS Strong EpCAM expression was observed in 77% of the tumors and in 85% of the corresponding lymph nodes. Of the primary tumors, 58% (n=74) presented a homogeneous intratumoral EpCAM expression while 42% were characterised by a heterogenous expression pattern. Tumors with high EpCAM expression at the invasive front were associated with significantly (p=0.03) higher proportion of lymph node metastases and lower median overall survival (p=0.001). Diffuse type tumors presented a significantly higher EpCAM expression at the invasion front compared with the tumor centre (p=0.036). Multivariate survival analysis identified high EpCAM expression at the invasive front as an independent prognostic factor.We observed a significant (p=0.001) correlation between high EpCAM expression and higher tumor cell proliferation. CONCLUSION High EpCAM expression associates with proliferation and progression of gastric cancer, especially in the diffuse type. Considering the discontenting results of the current adjuvant concepts for gastric cancer patients, EpCAM might be target in the adjuvant therapy of this malignant disease.
Collapse
Affiliation(s)
- Feride Kroepil
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Schnell U, Cirulli V, Giepmans BNG. EpCAM: structure and function in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:1989-2001. [PMID: 23618806 DOI: 10.1016/j.bbamem.2013.04.018] [Citation(s) in RCA: 209] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 04/12/2013] [Accepted: 04/16/2013] [Indexed: 12/14/2022]
Abstract
Injection of tumor cells in mice more than 30 years ago resulted in the discovery of an epithelial antigen, later defined as a cell adhesion molecule (EpCAM). Although EpCAM has since evoked significant interest as a target in cancer therapy, mechanistic insights on the functions of this glycoprotein have been emerging only very recently. This may have been caused by the multitude of functions attributed to the glycoprotein, its localization at different subcellular sites and complex posttranslational modifications. Here, we review how EpCAM modifies cell-cell contact adhesion strength and tissue plasticity, and how it regulates cell proliferation and differentiation. Major knowledge derived from human diseases will be highlighted: Mutant EpCAM that is absent from the cell surface leads to fatal intestinal abnormalities (congenital tufting enteropathy). EpCAM-mediated cell proliferation in cancer may result from signaling (i) via regulated intramembrane proteolysis and/or (ii) the localization and association with binding partners in specialized membrane microdomains. New insight in EpCAM signaling will help to develop optimized cancer therapies and open new avenues in the field of regenerative medicine.
Collapse
Affiliation(s)
- Ulrike Schnell
- Dept. of Cell Biology, University of Groningen, Groningen, The Netherlands
| | | | | |
Collapse
|
50
|
Miyake T, Nimura S, Hamada Y, Nabeshima K, Shinohara T, Tanaka S, Yamashita Y, Takeshita M, Iwasaki H. MK-1 expression in gastric carcinoma with liver metastasis. Jpn J Clin Oncol 2013; 43:377-82. [PMID: 23458716 DOI: 10.1093/jjco/hyt007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE The prognosis for gastric carcinoma patients with liver metastasis is very poor. This retrospective study investigated the prognostic significance of MK-1 expression in gastric carcinoma patients with liver metastasis. METHODS Immunohistochemical staining using monoclonal antibody FU-MK-1 against MK-1 antigen was performed on paraffin-embedded tissues from 64 gastric carcinoma patients with liver metastasis. We attempted to determine the presence of any relationship between pathological prognostic factors and the expression of MK-1 in 64 gastric carcinoma patients with liver metastasis. RESULTS MK-1 expression was found in 43 (67%) of 64 tumor samples. MK-1 expression was significantly higher in the intestinal type (73%) than in the diffuse type carcinoma (33%, P = 0.049). Multivariate analysis showed that MK-1 expression and lymph node metastasis were significant factors for overall survival. The difference between overall survival rates with positive or negative MK-1 expression was statistically significant as shown by Kaplan-Meier survival analysis (P < 0.0001; log-rank). In addition, the difference between cumulative disease-free survival rates with positive or negative MK-1 expression in gastric carcinoma patients with metachronous liver metastasis was statistically significant as well, as shown by Kaplan-Meier survival analysis (P = 0.0006; log-rank). CONCLUSIONS The prognostic significance of MK-1 expression as a biological tumor marker was demonstrated in a series of gastric carcinoma patients with liver metastasis. MK-1 positivity may be a reliable marker for predicting and taking measures to control liver metastasis after curative gastrectomy for gastric carcinoma.
Collapse
Affiliation(s)
- Toru Miyake
- Department of Pathology, Fukuoka University, Nanakuma, Fukuoka, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|