1
|
Balaji N, Kukal S, Bhat A, Pradhan N, Minocha S, Kumar S. A quartet of cancer stem cell niches in hepatocellular carcinoma. Cytokine Growth Factor Rev 2024; 79:39-51. [PMID: 39217065 DOI: 10.1016/j.cytogfr.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Hepatocellular Carcinoma (HCC), the most prevalent type of primary liver cancer, is known for its aggressive behavior and poor prognosis. The Cancer Stem Cell theory, which postulates the presence of a small population of self-renewing cells called Cancer Stem Cells (CSCs), provides insights into various clinical and molecular features of HCC such as tumor heterogeneity, metabolic adaptability, therapy resistance, and recurrence. These CSCs are nurtured in the tumor microenvironment (TME), where a mix of internal and external factors creates a tumor-supportive niche that is continuously evolving both spatially and temporally, thus enhancing the tumor's complexity. This review details the origins of hepatic CSCs (HCSCs) and the factors influencing their stem-like qualities. It highlights the reciprocal crosstalk between HCSCs and the TME (hypoxic, vascular, invasive, and immune niches), exploring the signaling pathways involved and how these interactions control the malignant traits of CSCs. Additionally, it discusses potential therapeutic approaches targeting the HCSC niche and their possible uses in clinical practice.
Collapse
Affiliation(s)
- Neha Balaji
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India
| | - Samiksha Kukal
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India
| | - Anjali Bhat
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India
| | - Nikita Pradhan
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India
| | - Shilpi Minocha
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India.
| | - Saran Kumar
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India.
| |
Collapse
|
2
|
Abdellateif MS, Zekri ARN. Stem cell therapy for hepatocellular carcinoma and end-stage liver disease. J Egypt Natl Canc Inst 2023; 35:35. [PMID: 37926787 DOI: 10.1186/s43046-023-00194-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 10/20/2023] [Indexed: 11/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major health problem worldwide, especially for patients who are suffering from end-stage liver disease (ESLD). The ESLD is considered a great challenge for clinicians due to the limited chance for liver transplantation, which is the only curative treatment for those patients. Stem cell-based therapy as a part of regenerative medicine represents a promising application for ESLD patients. Many clinical trials were performed to assess the utility of bone marrow-derived stem cells as a potential therapy for patients with liver diseases. The aim of the present study is to present and review the various types of stem cell-based therapy, including the mesenchymal stem cells (MSCs), BM-derived mononuclear cells (BM-MNCs), CD34 + hematopoietic stem cells (HSCs), induced pluripotent stem cells (iPSCs), and cancer stem cells.Though this type of therapy achieved promising results for the treatment of ESLD, however still there is a confounding data regarding its clinical application. A large body of evidence is highly required to evaluate the stem cell-based therapy after long-term follow-up, with respect to the incidence of toxicity, immunogenicity, and tumorigenesis that developed in many patients.
Collapse
Affiliation(s)
- Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11976, Egypt.
| | - Abdel-Rahman N Zekri
- Molecular Virology and Immunology Unit, Cancer Biology Department, NCI, Cairo University, Cairo, 11976, Egypt
| |
Collapse
|
3
|
Yang Q, Liang Y, Shi Y, Shang J, Huang X. The ALKBH5/SOX4 axis promotes liver cancer stem cell properties via activating the SHH signaling pathway. J Cancer Res Clin Oncol 2023; 149:15499-15510. [PMID: 37646828 DOI: 10.1007/s00432-023-05309-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023]
Abstract
Hepatocellular carcinoma (HCC), featured with high prevalence and poor prognosis, is the major cause of cancer-related deaths worldwide. As a subgroup of liver cancer cells capable of differentiation, tumorigenesis and self-renewal, liver cancer stem cells (LCSCs) serve as one of the reasons leading to HCC progression and therapeutic resistance. Therefore, in-depth exploration of novel molecular biomarkers related to LSCSs is of great necessity. In our study, we found that human AlkB homolog H5 (ALKBH5) expression was enriched in LCSCs, which could foster proliferation, invasion and migration of the HCC cells. Mechanically, ALKBH5 positively mediated the expression of SOX4 via demethylation, and SOX4 promoted SHH expression at the transcriptional level to activate sonic hedgehog (SHH) signaling pathway. Furthermore, exosomes derived from CD133+ HCC cells could transmit ALKBH5 into THP-1 cells, which might be associated with M2 polarization of macrophages. In summary, the ALKBH5/SOX4 axis plays a significant role in exacerbating LCSC properties via activating SHH signaling pathway, and ALKBH5 could be a critical effector related to macrophage M2 polarization. These findings might provide a promising new biomarker for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Qinyan Yang
- Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuxin Liang
- Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Shi
- Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jin Shang
- Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Xiaolun Huang
- Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
4
|
Vita F, Olaizola I, Amato F, Rae C, Marco S, Banales JM, Braconi C. Heterogeneity of Cholangiocarcinoma Immune Biology. Cells 2023; 12:cells12060846. [PMID: 36980187 PMCID: PMC10047186 DOI: 10.3390/cells12060846] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Cholangiocarcinomas (CCAs) are aggressive tumors arising along the biliary tract epithelium, whose incidence and mortality are increasing. CCAs are highly desmoplastic cancers characterized by a dense tumor microenvironment (TME), in which each single component plays a fundamental role in shaping CCA initiation, progression and resistance to therapies. The crosstalk between cancer cells and TME can affect the recruitment, infiltration and differentiation of immune cells. According to the stage of the disease and to intra- and inter-patient heterogeneity, TME may contribute to either protumoral or antitumoral activities. Therefore, a better understanding of the effect of each immune cell subtype may open the path to new personalized immune therapeutic strategies for the management of CCA. In this review, we describe the role of immune cells in CCA initiation and progression, and their crosstalk with both cancer-associated fibroblasts (CAFs) and the cancer-stem-cell-like (CSC) niche.
Collapse
Affiliation(s)
- Francesca Vita
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (F.V.); (F.A.); (C.R.); (S.M.)
- Department of Oncology, University of Turin, 10043 Turin, Italy
| | - Irene Olaizola
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute–Donostia University Hospital, University of the Basque Country (UPV/EHU), 20014 San Sebastian, Spain; (I.O.); (J.M.B.)
| | - Francesco Amato
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (F.V.); (F.A.); (C.R.); (S.M.)
| | - Colin Rae
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (F.V.); (F.A.); (C.R.); (S.M.)
| | - Sergi Marco
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (F.V.); (F.A.); (C.R.); (S.M.)
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute–Donostia University Hospital, University of the Basque Country (UPV/EHU), 20014 San Sebastian, Spain; (I.O.); (J.M.B.)
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, “Instituto de Salud Carlos III”), 28029 Madrid, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
| | - Chiara Braconi
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (F.V.); (F.A.); (C.R.); (S.M.)
- Beatson West of Scotland Cancer Centre, Glasgow G12 0YN, UK
- Correspondence:
| |
Collapse
|
5
|
Zhou XH, Li JR, Zheng TH, Chen H, Cai C, Ye SL, Gao B, Xue TC. Portal vein tumor thrombosis in hepatocellular carcinoma: molecular mechanism and therapy. Clin Exp Metastasis 2023; 40:5-32. [PMID: 36318440 DOI: 10.1007/s10585-022-10188-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022]
Abstract
Portal vein tumor thrombosis (PVTT), a common complication of advanced hepatocellular carcinoma (HCC), remains the bottleneck of the treatments. Liver cancer cells potentially experienced multi-steps during PVTT process, including cancer cells leave from cancer nest, migrate in extracellular matrix, invade the vascular barrier, and colonize in the portal vein. Accumulated evidences have revealed numerous of molecular mechanisms including genetic and epigenetic regulation, cancer stem cells, immunosuppressive microenvironment, hypoxia, et al. contributed to the PVTT formation. In this review, we discuss state-of-the-art PVTT research on the potential molecular mechanisms and experimental models. In addition, we summarize PVTT-associated clinical trials and current treatments for PVTT and suppose perspectives exploring the molecular mechanisms and improving PVTT-related treatment for the future.
Collapse
Affiliation(s)
- Xing-Hao Zhou
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, 136 Yi Xue Yuan Road, Shanghai, 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, China.,Department of Hepatic Oncology, Fudan University, Zhongshan Hospital, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China
| | - Jing-Ru Li
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, 136 Yi Xue Yuan Road, Shanghai, 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, China.,Department of Hepatic Oncology, Fudan University, Zhongshan Hospital, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China
| | - Tang-Hui Zheng
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, 136 Yi Xue Yuan Road, Shanghai, 200032, China.,Department of Hepatic Oncology, Xiamen Branch, Fudan University, Zhongshan Hospital, Xiamen, 361015, China
| | - Hong Chen
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, 136 Yi Xue Yuan Road, Shanghai, 200032, China.,Department of Hepatic Oncology, Xiamen Branch, Fudan University, Zhongshan Hospital, Xiamen, 361015, China
| | - Chen Cai
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, 136 Yi Xue Yuan Road, Shanghai, 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, China.,Department of Hepatic Oncology, Fudan University, Zhongshan Hospital, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China
| | - Sheng-Long Ye
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, 136 Yi Xue Yuan Road, Shanghai, 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, China.,Department of Hepatic Oncology, Fudan University, Zhongshan Hospital, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China
| | - Bo Gao
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai Medical College, Shanghai, 200032, China.
| | - Tong-Chun Xue
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, 136 Yi Xue Yuan Road, Shanghai, 200032, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, China. .,Department of Hepatic Oncology, Fudan University, Zhongshan Hospital, Shanghai, 200032, China. .,National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
6
|
Cancer Stem Cells in Hepatocellular Carcinoma: Intrinsic and Extrinsic Molecular Mechanisms in Stemness Regulation. Int J Mol Sci 2022; 23:ijms232012327. [PMID: 36293184 PMCID: PMC9604119 DOI: 10.3390/ijms232012327] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/18/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
Hepatocellular carcinoma (HCC) remains the most predominant type of liver cancer with an extremely poor prognosis due to its late diagnosis and high recurrence rate. One of the culprits for HCC recurrence and metastasis is the existence of cancer stem cells (CSCs), which are a small subset of cancer cells possessing robust stem cell properties within tumors. CSCs play crucial roles in tumor heterogeneity constitution, tumorigenesis, tumor relapse, metastasis, and resistance to anti-cancer therapies. Elucidation of how these CSCs maintain their stemness features is essential for the development of CSCs-based therapy. In this review, we summarize the present knowledge of intrinsic molecules and signaling pathways involved in hepatic CSCs, especially the CSC surface markers and associated signaling in regulating the stemness characteristics and the heterogeneous subpopulations within the CSC pool. In addition, we recapitulate the effects of crucial extrinsic cellular components in the tumor microenvironment, including stromal cells and immune cells, on the modulation of hepatic CSCs. Finally, we synopsize the currently valuable CSCs-targeted therapy strategies based on intervention in these intrinsic and extrinsic molecular mechanisms, in the hope of shedding light on better clinical management of HCC patients.
Collapse
|
7
|
Xie X, Chen J, Wo D, Ma E, Ning Y, Peng J, Zhu W, Ren DN. Babao Dan is a robust anti-tumor agent via inhibiting wnt/β-catenin activation and cancer cell stemness. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114449. [PMID: 34332067 DOI: 10.1016/j.jep.2021.114449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/15/2021] [Accepted: 07/22/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese Medicine (TCM) is being increasingly used worldwide due to its diverse efficacy and relatively low side effects. Babao Dan (BBD) is a well-known TCM formula that is currently used for the effective treatment of various cancers, however its underlying molecular mechanism remains unknown. AIM OF THE STUDY Tumor growth and tumor recurrence are characterized by two distinct populations of cells, namely the well-differentiated cancer cells composing the majority of tumor bulk, and cancer stem cells (CSCs) involved in tumor relapse, which are both strongly associated with excessive activation of Wnt/β-catenin signaling. Our study aims to elucidate the underlying molecular mechanisms associated with the anti-tumor proliferative effects of Babao Dan (BBD). MATERIALS AND METHODS We used a hepatoblastoma cell line HepG2 with stem cell-like traits that harbors a constitutively active mutant of β-catenin in order to study the anti-tumor ability of BBD via targeting Wnt/β-catenin signaling. RESULTS BBD robustly attenuated both the intrinsic and extrinsic activation of Wnt/β-catenin pathway in HepG2 hepatoblastoma cells, as well as Wnt target genes. Moreover, BBD significantly inhibited both the proliferation of well-differentiated cancer cells, as well as the stem-like property of CSCs as evidenced by EpCAM, a Wnt target gene and a novel marker of cancer cell stemness. In addition, mice administered with BBD using HepG2 cell line derived xenograft model had marked reductions in tumor size and weight, as well as significantly decreased expressions of Wnt target genes and cancer cell stemness. CONCLUSION Our findings elucidated the underlying molecular mechanisms associated with the robust anti-tumor effects of BBD via potent inhibition of Wnt/β-catenin signaling, and implicate its use in the clinical treatment of cancers.
Collapse
Affiliation(s)
- Xinxin Xie
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Jinxiao Chen
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Da Wo
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - En Ma
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Yongling Ning
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Jun Peng
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Weidong Zhu
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Dan-Ni Ren
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China.
| |
Collapse
|
8
|
DT389-YP7, a Recombinant Immunotoxin against Glypican-3 That Inhibits Hepatocellular Cancer Cells: An In Vitro Study. Toxins (Basel) 2021; 13:toxins13110749. [PMID: 34822533 PMCID: PMC8617615 DOI: 10.3390/toxins13110749] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/09/2021] [Accepted: 10/13/2021] [Indexed: 01/02/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the high-metastatic types of cancer, and metastasis occurs in one-third of patients with HCC. To maintain the effectiveness of drug compounds on cancer cells and minimize their side effects on normal cells, it is important to use new approaches for overcoming malignancies. Immunotoxins (ITs), an example of such a new approach, are protein-structured compounds consisting of toxic and binding moieties which can specifically bind to cancer cells and efficiently induce cell death. Here, we design and scrutinize a novel immunotoxin against an oncofetal marker on HCC cells. We applied a truncated diphtheria toxin (DT389) without binding domain as a toxin moiety to be fused with a humanized YP7 scFv against a high-expressed Glypican-3 (GPC3) antigen on the surface of HCC cells. Cytotoxic effects of this IT were investigated on HepG2 (GPC3+) and SkBr3 (GPC3−) cell lines as positive- and negative-expressed GPC3 antigens. The dissociation constant (Kd) was calculated 11.39 nM and 18.02 nM for IT and YP7 scfv, respectively, whereas only IT showed toxic effects on the HepG2 cell line, and decreased cell viability (IC50 = 848.2 ng/mL). Changing morphology (up to 85%), cell cycle arrest at G2 phase (up to 13%), increasing intracellular reactive oxygen species (ROSs) (up to 50%), inducing apoptosis (up to 38% for apoptosis and 23% for necrosis), and an almost complete inhibition of cell movement were other effects of immunotoxin treatment on HepG2 cells, not on SkBr3 cell line. These promising results reveal that this new recombinant immunotoxin can be considered as an option as an HCC inhibitor. However, more extensive studies are needed to accomplish this concept.
Collapse
|
9
|
Immunotoxins Immunotherapy against Hepatocellular Carcinoma: A Promising Prospect. Toxins (Basel) 2021; 13:toxins13100719. [PMID: 34679012 PMCID: PMC8538445 DOI: 10.3390/toxins13100719] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/02/2021] [Accepted: 10/03/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers in the world. Therefore, fighting against such cancer is reasonable. Chemotherapy drugs are sometimes inefficient and often accompanied by undesirable side effects for patients. On the other hand, the emergence of chemoresistant HCC emphasizes the need for a new high-efficiency treatment strategy. Immunotoxins are armed and rigorous targeting agents that can purposefully kill cancer cells. Unlike traditional chemotherapeutics, immunotoxins because of targeted toxicity, insignificant cross-resistance, easy production, and other favorable properties can be ideal candidates against HCC. In this review, the characteristics of proper HCC-specific biomarkers for immunotoxin targeting were dissected. After that, the first to last immunotoxins developed for the treatment of liver cancer were discussed. So, by reviewing the strengths and weaknesses of these immunotoxins, we attempted to provide keynotes for designing an optimal immunotoxin against HCC.
Collapse
|
10
|
Kwon SH, Faruque HA, Kee H, Kim E, Park S. Exosome-based hybrid nanostructures for enhanced tumor targeting and hyperthermia therapy. Colloids Surf B Biointerfaces 2021; 205:111915. [PMID: 34130212 DOI: 10.1016/j.colsurfb.2021.111915] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/29/2021] [Accepted: 06/06/2021] [Indexed: 02/06/2023]
Abstract
Recently, natural exosomes have attracted attention as an ideal drug carrier to overcome the limitations of existing drug delivery systems which are toxicity induction and low cancer-targeting performance. In this study, we propose an exosome-based hybrid nanostructure (EHN) with improved targeting ability and therapeutic efficacy against colorectal cancer by using exosomes isolated from the tumor cell line as a drug carrier. The proposed EHN can have high biocompatibility by using exosomes, a biologically derived material, and show improved targeting performance by adding a tumor-targeting ligand (folic acid). In addition, the proposed EHN is capable of chemotherapy because doxorubicin, an anticancer drug, is encapsulated by the exosome with high efficiency, and it can induce hyperthermia therapy because of the magnetic nanoparticles (MNPs) attached to the surface of exosomes. Through in vitro and in vivo experiments using a xenograft tumor mouse model, it was confirmed that the proposed EHN could exhibit increased apoptosis and excellent tumor growth inhibition ability. Therefore, the proposed EHN is expected to overcome the limitations of existing drug delivery systems and be utilized as an effective drug delivery system in cancer treatment.
Collapse
Affiliation(s)
- Su-Hyun Kwon
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea; Department of Chemistry, College of Science, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Hasan Al Faruque
- Companion Diagnostics and Medical Technology Research Group, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Hyeonwoo Kee
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Eunjoo Kim
- Companion Diagnostics and Medical Technology Research Group, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
| | - Sukho Park
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
| |
Collapse
|
11
|
Jung J, Joe H, Ha K, Lim JM, Kim HG. Biomedical Entity Explorer: A Web Server for Biomedical Entity Exploration. J Comput Biol 2021; 28:619-628. [PMID: 34081565 DOI: 10.1089/cmb.2020.0364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Biomedical Entity Explorer (BEE) is a web server that can search for biomedical entities from a database of six biomedical entity types (gene, miRNA, drug, disease, single nucleotide polymorphism [SNP], pathway) and their gene associations. The search results can be explored using intersections, unions, and negations. BEE has integrated biomedical entities from 16 databases (Ensemble, PharmGKB, Genetic Home Reference, Tarbase, Mirbase, NCI Thesaurus, DisGeNET, Linked life data, UMLS, GSEA MsigDB, Reactome, KEGG, Gene Ontology, HGVD, SNPedia, and dbSNP) based on their gene associations and built a database with their synonyms, descriptions, and links containing individual details. Users can enter the keyword of one or more entities and select the type of entity for which they want to know the relationship for and by using set operations such as union, negation, and intersection, they can navigate the search results more clearly. We believe that BEE will not only be useful for biologists querying for complex associations between entities, but can also be a good starting point for general users searching for biomedical entities. BEE is accessible at (http://bike-bee.snu.ac.kr).
Collapse
Affiliation(s)
- Jinuk Jung
- Biomedical Knowledge Engineering Laboratory, Seoul National University School of Dentistry, Seoul, Korea.,Alopax-Algo, Co. Ltd, Seoul, Korea
| | - Hyunwhan Joe
- Biomedical Knowledge Engineering Laboratory, Seoul National University School of Dentistry, Seoul, Korea
| | - Kyungsik Ha
- Dental Research Institute, Seoul National University, Seoul, Korea
| | - Jin-Muk Lim
- Biomedical Knowledge Engineering Laboratory, Seoul National University School of Dentistry, Seoul, Korea.,Alopax-Algo, Co. Ltd, Seoul, Korea
| | - Hong-Gee Kim
- Biomedical Knowledge Engineering Laboratory, Seoul National University School of Dentistry, Seoul, Korea.,Dental Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
12
|
PLGA nanoparticles containing α-fetoprotein siRNA induce apoptosis and enhance the cytotoxic effects of doxorubicin in human liver cancer cell line. Biochem Biophys Res Commun 2021; 553:191-197. [PMID: 33774221 DOI: 10.1016/j.bbrc.2021.03.086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/15/2021] [Indexed: 01/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers and is a leading cause of death. Delivery of therapeutic molecules, e.g., siRNA, to HCC cells could potentially be an alternative treatment for HCC. In this study, the siRNA targeting α-fetoprotein (AFP) mRNA was found to specifically induce apoptosis and significant cell death in HepG2 cells. It also enhanced the cytotoxic effects of doxorubicin by about two-fold, making it the candidate therapeutic molecule for HCC treatment. To deliver the siRNAs into HCC cells, the AFP siRNAs were loaded into the nanoparticles based on poly (lactic-co-glycolic) acid (PLGA). These nanoparticles induced apoptosis in HepG2 cells and synergistically increased the cytotoxicity of doxorubicin. In summary, the delivery of the AFP siRNA-loaded PLGA nanoparticles in combination with doxorubicin could be a very promising approach for the treatment of HCC.
Collapse
|
13
|
Patel K, Lamm R, Altshuler P, Dang H, Shah AP. Hepatocellular Carcinoma-The Influence of Immunoanatomy and the Role of Immunotherapy. Int J Mol Sci 2020; 21:ijms21186757. [PMID: 32942580 PMCID: PMC7555667 DOI: 10.3390/ijms21186757] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related morbidity and mortality worldwide. Most patients are diagnosed with advanced disease, limiting their options for treatment. While current treatments are adequate for lower staged disease, available systemic treatments are limited, with marginal benefit at best. Chimeric antigen receptor (CAR) T cell therapy, effective in treating liquid tumors such as B-cell lymphoma, presents a potentially promising treatment option for advanced HCC. However, new challenges specific to solid tumors, such as tumor immunoanatomy or the immune cell presence and position anatomically and the tumor microenvironment, need to be defined and overcome. Immunotherapy currently in use must be re-engineered and re-envisioned to treat HCC with the hopes of ushering in an answer to advanced stage solid tumor disease processes. Future therapy options must address the uniqueness of the tumors under the umbrella of HCC. This review strives to summarize HCC, its staging system, current therapy and immunotherapy medications currently being utilized or studied in the treatment of HCC with the hopes of highlighting what is being done and suggesting what needs to be done in the future to champion this therapy as an effective option.
Collapse
Affiliation(s)
- Keyur Patel
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19144, USA; (K.P.); (R.L.); (P.A.)
| | - Ryan Lamm
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19144, USA; (K.P.); (R.L.); (P.A.)
| | - Peter Altshuler
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19144, USA; (K.P.); (R.L.); (P.A.)
| | - Hien Dang
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19144, USA; (K.P.); (R.L.); (P.A.)
- Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
- Correspondence: (H.D.); (A.P.S.)
| | - Ashesh P. Shah
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19144, USA; (K.P.); (R.L.); (P.A.)
- Correspondence: (H.D.); (A.P.S.)
| |
Collapse
|
14
|
Dal Bo M, De Mattia E, Baboci L, Mezzalira S, Cecchin E, Assaraf YG, Toffoli G. New insights into the pharmacological, immunological, and CAR-T-cell approaches in the treatment of hepatocellular carcinoma. Drug Resist Updat 2020; 51:100702. [DOI: 10.1016/j.drup.2020.100702] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 02/07/2023]
|
15
|
Liu YC, Yeh CT, Lin KH. Cancer Stem Cell Functions in Hepatocellular Carcinoma and Comprehensive Therapeutic Strategies. Cells 2020; 9:cells9061331. [PMID: 32466488 PMCID: PMC7349579 DOI: 10.3390/cells9061331] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a significant cause of cancer-related mortality owing to resistance to traditional treatments and tumor recurrence after therapy, which leads to poor therapeutic outcomes. Cancer stem cells (CSC) are a small subset of tumor cells with the capability to influence self-renewal, differentiation, and tumorigenesis. A number of surface markers for liver cancer stem cell (LCSC) subpopulations (EpCAM, CD133, CD44, CD13, CD90, OV-6, CD47, and side populations) in HCC have been identified. LCSCs play critical roles in regulating HCC stemness, self-renewal, tumorigenicity, metastasis, recurrence, and therapeutic resistance via genetic mutations, epigenetic disruption, signaling pathway dysregulation, or alterations microenvironment. Accumulating studies have shown that biomarkers for LCSCs contribute to diagnosis and prognosis prediction of HCC, supporting their utility in clinical management and development of therapeutic strategies. Preclinical and clinical analyses of therapeutic approaches for HCC using small molecule inhibitors, oncolytic measles viruses, and anti-surface marker antibodies have demonstrated selective, efficient, and safe targeting of LCSC populations. The current review focuses on recent reports on the influence of LCSCs on HCC stemness, tumorigenesis, and multiple drug resistance (MDR), along with LCSC-targeted therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Yu-Chin Liu
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan;
- Department of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
| | - Kwang-Huei Lin
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan;
- Department of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Correspondence: ; Tel./Fax: +886-3-211-8263
| |
Collapse
|
16
|
Zhou C, Sun J, Zheng Z, Weng J, Atyah M, Zhou Q, Chen W, Zhang Y, Huang J, Yin Y, Mao H, Zhang Z, Yi Y, Ren N. High RPS11 level in hepatocellular carcinoma associates with poor prognosis after curative resection. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:466. [PMID: 32395510 PMCID: PMC7210141 DOI: 10.21037/atm.2020.03.92] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Ribosomal protein S11 (RPS11), a member of ribosomal protein family, is reported to overexpress in diverse malignancies and correlates with tumor recurrence. However, our current knowledge on RPS11 in hepatocellular carcinoma (HCC) remains limited. In this study, we are going to explore the potential prognostic value of RPS11 in HCC patients after curative resection. Methods Immunohistochemistry (IHC) was performed to evaluate RPS11 expression on tissue microarrays in training cohort comprising 182 HCC patients and validation cohort enrolling 90 HCC patients in Zhongshan Hospital, Fudan University. Western blot and quantitative reverse transcription PCR (qRT-PCR) were also used to determine the expression level of RPS11 in liver cell lines. Two nomograms, calibration curves and decision curve analysis (DCA) were further performed to assess the performance of RPS11 level in predicting clinical outcomes of HCC patients. Additionally, single-sample gene-set enrichment analysis (ssGSEA) was conducted in TCGA liver cancer database to investigate the potential biological pathways involved in RPS11. Results Both increased mRNA and protein levels of RPS11 were observed in most HCC cell lines when compared to the normal hepatocytes, and high tumor RPS11 level was associated with shorter overall survival (OS) and recurrence-free survival (RFS) of HCC patients after curative resection. Univariate and multivariate analysis indicated that RPS11 was an independent prognostic factor in HCC. Two nomograms, calibration and DCA curves were further established and displayed a superior prognostic accuracy of OS and RFS, and showed more clinical benefits than traditional staging systems in HCC. Furthermore, several pathways and molecules related to tumor resistance, survival and recurrence were enriched in high RPS11 expression by ssGSEA. Conclusions Tumorous RPS11 acts as a potential prognostic biomarker for HCC patients who received curative resection.
Collapse
Affiliation(s)
- Chenhao Zhou
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Institute of Fudan Minhang Academic Health System, Minhang Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jialei Sun
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China.,Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhiyuan Zheng
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University and Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Jialei Weng
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Manar Atyah
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Qiang Zhou
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Wanyong Chen
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China.,Institute of Fudan Minhang Academic Health System, Minhang Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuan Zhang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jiaxing Huang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yirui Yin
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Huarong Mao
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China.,Institute of Fudan Minhang Academic Health System, Minhang Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ziping Zhang
- Institute of Fudan Minhang Academic Health System, Minhang Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yong Yi
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Ning Ren
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China.,Institute of Fudan Minhang Academic Health System, Minhang Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Abdelgawad IA. Epithelial Cell Adhesion Molecule mRNA Can be a Potential Marker to Predict Metastasis in Hepatocellular Carcinoma Patients. Asian Pac J Cancer Prev 2020; 21:861-866. [PMID: 32212818 PMCID: PMC7437321 DOI: 10.31557/apjcp.2020.21.3.861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Epithelial cell adhesion molecule [EpCAM] is a surface marker of cancer stem cells that can maintain the capacity for malignant proliferation, invasion, metastasis, and tumor recurrence; hence its detection among hepatocellular carcinoma [HCC] patients may be an important prognostic factor. The aim of this study was to detect EpCAM mRNA expression in the whole blood of HCC patients and normal control subjects to elucidate its clinico-pathological significance among patients with HCC. METHODS This study was conducted on 74 newly diagnosed HCC patients and forty normal control subjects. Both groups were subjected to the detection of EpCAM mRNA in the whole blood using reverse transcriptase polymerase chain reaction [RT- PCR] technique. EpCAM expression was compared with some of the established prognostic factors of HCC. RESULTS EpCAM was detected in 17.5% of the HCC cases and was not expressed in any of the normal control subjects. EpCAM positive cases showed higher serum levels of alpha- feto protein [AFP] and carcinoembryonic antigen [CEA]. Prevalence of EpCAM positivity gave significant results with distant metastasis, lymph node metastasis, and portal vein thrombosis. CONCLUSION EpCAM proved high specificity among HCC patients and its expression was associated with metastasis and portal vein thrombosis. Higher serum levels of CEA among the EpCAM positive patients may attract the attention to a subgroup of HCC patients who are more liable to develop metastasis.<br />.
Collapse
Affiliation(s)
- Iman A Abdelgawad
- Department of Clinical Pathology, NCI, Cairo University, Cairo, Egypt
| |
Collapse
|
18
|
Busato D, Mossenta M, Baboci L, Di Cintio F, Toffoli G, Dal Bo M. Novel immunotherapeutic approaches for hepatocellular carcinoma treatment. Expert Rev Clin Pharmacol 2019; 12:453-470. [PMID: 30907177 DOI: 10.1080/17512433.2019.1598859] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The introduction of immune checkpoint inhibitors has been lately proposed for the treatment of hepatocellular carcinoma (HCC) with respect to other cancer types. Several immunotherapeutic approaches are now under evaluation for HCC treatment including: i) antibodies acting as immune checkpoint inhibitors; ii) antibodies targeting specific tumor-associated antigens; iii) chimeric antigen receptor redirected T (CAR-T) cells targeting specific tumor-associated antigens; iv) vaccination strategies with tumor-specific epitopes. Areas covered: The review provides a wide description of the clinical trials investigating the efficacy of the main immunotherapeutic approaches proposed for the treatment of patients affected by HCC. Expert opinion: The balancing between immunostimulative and immunosuppressive factors in the context of HCC tumor microenvironment results in heterogeneous response rates to immunotherapeutic approaches such as checkpoint inhibitors, among HCC patients. In this context, it becomes crucial the identification of predictive factors determining the treatment response. A multiple approach using different biomarkers could be useful to identify the subgroup of HCC patients responsive to the treatment with a checkpoint inhibitor (as an example, nivolumab) as single agent, and to identify those patients in which other treatment regimens, such as the combination with sorafenib, or with locoregional therapies, could be more effective.
Collapse
Affiliation(s)
- Davide Busato
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Monica Mossenta
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Lorena Baboci
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| | - Federica Di Cintio
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Giuseppe Toffoli
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| | - Michele Dal Bo
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| |
Collapse
|
19
|
Wang N, Wang S, Li MY, Hu BG, Liu LP, Yang SL, Yang S, Gong Z, Lai PBS, Chen GG. Cancer stem cells in hepatocellular carcinoma: an overview and promising therapeutic strategies. Ther Adv Med Oncol 2018; 10:1758835918816287. [PMID: 30622654 PMCID: PMC6304707 DOI: 10.1177/1758835918816287] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/06/2018] [Indexed: 12/12/2022] Open
Abstract
The poor clinical outcome of hepatocellular carcinoma (HCC) patients is ascribed to the resistance of HCC cells to traditional treatments and tumor recurrence after curative therapies. Cancer stem cells (CSCs) have been identified as a small subset of cancer cells which have high capacity for self-renewal, differentiation and tumorigenesis. Recent advances in the field of liver CSCs (LCSCs) have enabled the identification of CSC surface markers and the isolation of CSC subpopulations from HCC cells. Given their central role in cancer initiation, metastasis, recurrence and therapeutic resistance, LCSCs constitute a therapeutic opportunity to achieve cure and prevent relapse of HCC. Thus, it is necessary to develop therapeutic strategies to selectively and efficiently target LCSCs. Small molecular inhibitors targeting the core stemness signaling pathways have been actively pursued and evaluated in preclinical and clinical studies. Other alternative therapeutic strategies include targeting LCSC surface markers, interrupting the CSC microenvironment, and altering the epigenetic state. In this review, we summarize the properties of CSCs in HCC and discuss novel therapeutic strategies that can be used to target LCSCs.
Collapse
Affiliation(s)
- Nuozhou Wang
- Department of Surgery, The Chinese University of
Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR,
China
| | - Shanshan Wang
- Department of Otorhinolaryngology, Head and Neck
Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of
Wales Hospital, Hong Kong, China
| | - Ming-Yue Li
- Department of Surgery, Faculty of Medicine, The
Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong,
China
- Shenzhen Research Institute, The Chinese
University of Hong Kong, Shenzhen, Guangdong, China
| | - Bao-guang Hu
- Department of Gastrointestinal Surgery, The
Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong,
China
| | - Li-ping Liu
- Department of Hepatobiliary and Pancreas
Surgery, The Second Clinical Medical College of Jinan University (Shenzhen
People’s Hospital), Shenzhen, Guangdong Province, China
| | - Sheng-li Yang
- Cancer Center, Union Hospital, Tongji Medical
College, Huazhong University of Science and Technology, Wuhan, China
| | - Shucai Yang
- Department of Clinical Laboratory, Pingshan
District People’s Hospital of Shenzhen, Shenzhen, Guangdong Province,
China
| | - Zhongqin Gong
- Department of Surgery, The Chinese University of
Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR,
China
| | - Paul B. S. Lai
- Department of Surgery, The Chinese University
of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong
SAR, China
| | - George G. Chen
- Department of Surgery, The Chinese University
of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong
SAR, China
- Shenzhen Research Institute, The Chinese
University of Hong Kong, Shenzhen, Guangdong, China
| |
Collapse
|
20
|
Tanaka S. Precision medicine based on surgical oncology in the era of genome-scale analysis and genome editing technology. Ann Gastroenterol Surg 2018; 2:106-115. [PMID: 29863171 PMCID: PMC5881373 DOI: 10.1002/ags3.12059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/21/2017] [Indexed: 12/25/2022] Open
Abstract
Accumulated evidence suggests that multiple molecular and cellular interactions promote cancer evolution in vivo. Surgical oncology is of growing significance to a comprehensive understanding of the malignant diseases for therapeutic application. We have analyzed more than 1000 clinical samples from surgically resected tissue to identify molecular biomarkers and therapeutic targets for advanced malignancies. Cancer stemness and mitotic instability were then determined as the essential predictors of aggressive phenotype with poor prognosis. Recently, whole genome/exome sequencing showed a mutational landscape underlying phenotype heterogeneity in caners. In addition, integrated genomic, epigenomic, transcriptomic, metabolic, proteomic and phenomic analyses elucidated several molecular subtypes that cluster in liver, pancreatic, biliary, esophageal and gastroenterological cancers. Identification of each molecular subtype is expected to realize the precise medicine targeting subtype‐specific molecules; however, there are obstacle limitations to determine matching druggable targets or synthetic lethal interactions. Current breakthroughs in genome editing technology can provide us with unprecedented opportunity to recapitulate subtype‐specific pathophysiology in vitro and in vivo. Given a great potential, on‐demand editing system can design actionable strategy and revolutionize precision cancer medicine based on surgical oncology.
Collapse
Affiliation(s)
- Shinji Tanaka
- Department of Molecular Oncology Tokyo Medical and Dental University Tokyo Japan
| |
Collapse
|
21
|
The Recent Advances on Liver Cancer Stem Cells: Biomarkers, Separation, and Therapy. Anal Cell Pathol (Amst) 2017; 2017:5108653. [PMID: 28819584 PMCID: PMC5551471 DOI: 10.1155/2017/5108653] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/05/2017] [Indexed: 12/18/2022] Open
Abstract
As the third major reason of mortality related to cancer in the world, liver cancer is also the fifth most frequent cancer. Unluckily, a majority of patients succumb and relapse though many progresses have been made in detection and therapy of liver cancer. It has been put forward that in liver cancer, cancer stem cells (CSCs) hold main responsibility for the formation, invasion, metastasis, and recurrence of tumor. Strategies that are intended to target liver CSCs are playing a more and more significant role in supervising the development of liver cancer treatment and assessing new therapeutic methods. Herein, a brief review about molecule markers, signal pathways, separation, and treatment on liver cancer stem cells (LCSCs) is provided in this paper.
Collapse
|
22
|
|
23
|
Glypican-3 Targeting Immunotoxins for the Treatment of Liver Cancer. Toxins (Basel) 2016; 8:toxins8100274. [PMID: 27669301 PMCID: PMC5086635 DOI: 10.3390/toxins8100274] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/13/2016] [Accepted: 09/14/2016] [Indexed: 12/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer, yet no effective therapeutics exist. This review provides an overview of the recent development of recombinant immunotoxins for the treatment of glypican-3 (GPC3) expressing HCC. GPC3 is a cell surface heparan sulfate proteoglycan that is overexpressed in HCC, but is absent from normal adult human tissues. Treatment of HCC with anti-GPC3 immunotoxins represents a new therapeutic option. Using phage display and hybridoma technologies, three high affinity antibodies (HN3, HS20 and YP7) have been generated against GPC3. Two of these antibodies (HN3 and HS20) have demonstrated the ability to inhibit Wnt/Yap signaling, leading to a reduction in liver cancer cell proliferation. By combining the HN3 antibody capable of inhibiting Wnt/Yap signaling with the protein synthesis inhibitory domain of the Pseudomonas exotoxin, a recombinant immunotoxin that exhibits a dual inhibitory mechanism was generated. This immunotoxin was found to be highly effective in the treatment of human HCCs in mouse xenograft models. Engineering of the toxin fragment to reduce the level of immunogenicity is currently being explored. The development of immunotoxins provides opportunities for novel liver cancer therapies.
Collapse
|
24
|
Akahoshi K, Tanaka S, Mogushi K, Shimada S, Matsumura S, Akiyama Y, Aihara A, Mitsunori Y, Ban D, Ochiai T, Kudo A, Arii S, Tanabe M. Expression of connective tissue growth factor in the livers of non-viral hepatocellular carcinoma patients with metabolic risk factors. J Gastroenterol 2016; 51:910-22. [PMID: 26739296 DOI: 10.1007/s00535-015-1159-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/21/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND The incidence of hepatocellular carcinoma (HCC) associated with metabolic risk factors, such as diabetes and obesity, has been increasing. However, the underlying mechanism that links these diseases remains unclear. METHODS We performed genome-wide expression analysis of human liver tissues of non-viral HCC patients with or without metabolic risk factors. The upregulated genes that associated with diabetes and obesity were investigated by in vitro and in vivo experiments, and immunohistochemistry of human liver tissues was performed. RESULTS Among the upregulated genes, connective tissue growth factor (CTGF) expression was induced to a greater extent by combined glucose and insulin administration to human hepatoma cells. Genome-wide expression analysis revealed upregulation of a chemokine network in CTGF-overexpressing hepatoma cells, which displayed an increased ability to induce in vitro activation of macrophages, and in vivo infiltration of liver macrophages. Immunohistochemistry of human liver tissues validated the correlations between CTGF expression and diabetes or obesity as well as activation of liver macrophages in patients with non-viral HCC. Recurrence-free survival was significantly poorer in the CTGF-positive patients compared with the CTGF-negative patients (p = 0.002). Multivariate analysis determined that CTGF expression (HR 2.361; 95 % CI 1.195-4.665; p = 0.013) and vascular invasion (HR 2.367; 95 % CI 1.270-4.410; p = 0.007) were independent prognostic factors for recurrence of non-viral HCC. CONCLUSIONS Our data suggest that CTGF could be involved in oncogenic pathways promoting non-viral HCC associated with metabolic risk factors via induction of liver inflammation and is expected to be a novel HCC risk biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Keiichi Akahoshi
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.,Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan. .,Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Kaoru Mogushi
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Shu Shimada
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Satoshi Matsumura
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshimitsu Akiyama
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Arihiro Aihara
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Mitsunori
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Ban
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takanori Ochiai
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Kudo
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigeki Arii
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
25
|
Park SY, Bae JS, Cha EJ, Chu HH, Sohn JS, Moon WS. Nuclear EpICD expression and its role in hepatocellular carcinoma. Oncol Rep 2016; 36:197-204. [PMID: 27176150 DOI: 10.3892/or.2016.4789] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 02/18/2016] [Indexed: 11/06/2022] Open
Abstract
Regulated intramembrane proteolysis of epithelial cell adhesion molecule (EpCAM) results in shedding of the extracellular domain (EpEX) and release of the intra-cellular domain (EpICD) into the cytoplasm. Released EpICD associates with FHL2, β-catenin and Lef-1 to form a nuclear complex and triggers oncogenic signaling. This study was conducted to examine the nuclear expression of EpICD in hepatocellular carcinoma (HCC) and to assess the role of EpICD in HCC. EpICD immunoexpression was examined in 100 cases of HCC using tissue microarrays and correlated with clinicopathological parameters. We also examined the role of EpICD in HCC using EpICD cDNA transfected HCC cell line and EpCAM silenced HCC cell line by small interfering RNA (siRNA). Nuclear expression of EpICD was observed in 19 of 100 (19%) cases. Nuclear expression of EpICD significantly correlated with nuclear expression of β-catenin, and Ki-67 labeling index. In addition, nuclear expression of EpICD was associated with higher histologic grade and advanced T category. Forced overexpression of EpICD in the HCC cell significantly increased the cell proliferation, migration and invasion. The overexpression of EpICD also increased the expression levels of the active form of β-catenin and c-myc and cyclin D1. In contrast, downregulation of EpCAM by siRNA decreased the cell proliferation, migration, invasion and the expression of active form of β-catenin, c-myc and cyclin D1. Our present data suggest that EpICD plays important roles in HCC progression by modulating expression of target genes of EpCAM.
Collapse
Affiliation(s)
- Shin Young Park
- Department of Pathology, Konyang University College of Medicine, Daejeon 35365, Republic of Korea
| | - Jun Sang Bae
- Department of Pathology, Chonbuk National University, Medical School Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju 561-756, Republic of Korea
| | - Eun Jung Cha
- Department of Pathology, Konyang University College of Medicine, Daejeon 35365, Republic of Korea
| | - Hyun Hee Chu
- Department of Pathology, Konyang University College of Medicine, Daejeon 35365, Republic of Korea
| | - Jang Sihn Sohn
- Department of Pathology, Konyang University College of Medicine, Daejeon 35365, Republic of Korea
| | - Woo Sung Moon
- Department of Pathology, Chonbuk National University, Medical School Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju 561-756, Republic of Korea
| |
Collapse
|
26
|
The Therapeutic Targets of miRNA in Hepatic Cancer Stem Cells. Stem Cells Int 2016; 2016:1065230. [PMID: 27118975 PMCID: PMC4826947 DOI: 10.1155/2016/1065230] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/23/2016] [Accepted: 03/14/2016] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide malignancy and the third leading cause of cancer death in patients. Several studies demonstrated that hepatic cancer stem cells (HCSCs), also called tumor-initiating cells, are involved in regulation of HCC initiation, tumor progression, metastasis development, and drug resistance. Despite the extensive research, the underlying mechanisms by which HCSCs are regulated remain still unclear. MicroRNAs (miRNAs) are able to regulate a lot of biological processes such as self-renewal and pluripotency of HCSCs, representing a new promising strategy for treatment of HCC chemotherapy-resistant tumors. In this review, we synthesize the latest findings on therapeutic regulation of HCSCs by miRNAs, in order to highlight the perspective of novel miRNA-based anticancer therapies for HCC treatment.
Collapse
|
27
|
Chiba T, Iwama A, Yokosuka O. Cancer stem cells in hepatocellular carcinoma: Therapeutic implications based on stem cell biology. Hepatol Res 2016; 46:50-7. [PMID: 26123821 DOI: 10.1111/hepr.12548] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/13/2015] [Accepted: 06/22/2015] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer and the third most frequent cause of cancer-related death worldwide. Despite advances in its diagnosis and treatment, the prognosis of patients with advanced HCC remains unfavorable. Recent advances in stem cell biology and associated technologies have enabled the identification of minor components of tumorigenic cells, termed cancer stem cells (CSC) or tumor-initiating cells, in cancers such as HCC. Furthermore, because CSC play a central role in tumor development, metastasis and recurrence, they are considered to be a therapeutic target in cancer treatment. Hepatic CSC have been successfully identified using functional and cell surface markers. The analysis of purified hepatic CSC has revealed the molecular machinery and signaling pathways involved in their maintenance. In addition, epigenetic transcriptional regulation has been shown to be important in the development and maintenance of CSC. Although inhibitors of CSC show promise as CSC-targeting drugs, novel therapeutic approaches for the eradication of CSC are yet to be established. In this review, we describe recent progress in hepatic CSC research and provide a perspective on the available therapeutic approaches based on stem cell biology.
Collapse
Affiliation(s)
- Tetsuhiro Chiba
- Departments of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Iwama
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Osamu Yokosuka
- Departments of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
28
|
KATSUTA ERIKO, TANAKA SHINJI, MOGUSHI KAORU, SHIMADA SHU, AKIYAMA YOSHIMITSU, AIHARA ARIHIRO, MATSUMURA SATOSHI, MITSUNORI YUSUKE, BAN DAISUKE, OCHIAI TAKANORI, KUDO ATSUSHI, FUKAMACHI HIROSHI, TANAKA HIROSHI, NAKAYAMA KOH, ARII SHIGEKI, TANABE MINORU. CD73 as a therapeutic target for pancreatic neuroendocrine tumor stem cells. Int J Oncol 2015; 48:657-69. [DOI: 10.3892/ijo.2015.3299] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Accepted: 11/05/2015] [Indexed: 11/06/2022] Open
|
29
|
Construction, expression, and characterization of a recombinant immunotoxin targeting EpCAM. Mediators Inflamm 2015; 2015:460264. [PMID: 25960617 PMCID: PMC4415474 DOI: 10.1155/2015/460264] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/04/2014] [Indexed: 11/30/2022] Open
Abstract
Epithelial cell adhesion molecule (EpCAM) is a type I transmembrane glycoprotein overexpressed in human epithelioma but with relatively low expression in normal epithelial tissues. To exploit this differential expression pattern for targeted cancer therapy, an EpCAM-targeted immunotoxin was developed and its antitumor activity was investigated in vitro. An immunotoxin (scFv2A9-PE or APE) was constructed by genetically fusing a truncated form (PE38KDEL) of Pseudomonas aeruginosa exotoxin with an anti-EpCAM single-chain variable fragment (scFv). ELISA and flow cytometry were performed to verify immunotoxin (scFv2A9-PE or APE) antigen-binding activity with EpCAM. Cytotoxicity was measured by MTT assay. Confocal microscopy was used to observe its cellular localization. The results of ELISA and flow cytometry revealed that the immunotoxin efficiently recognized recombinant and natural EpCAM. Its antigen-binding activity was relatively lower than 2A9. MTT assay confirmed potent reduction in EpCAM-positive HHCC (human hepatocellular carcinoma) cell viability (IC50 50 pM). Immunofluorescence revealed that the immunotoxin localized to endoplasmic reticulum 24 h later. In conclusion, we described the development of an EpCAM-targeted immunotoxin with potent activity against tumor cells, which may lay the foundation for future development of therapeutic antibody for the treatment of EpCAM-positive tumors.
Collapse
|
30
|
Tanaka S. Cancer stem cells as therapeutic targets of hepato-biliary-pancreatic cancers. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2015; 22:531-7. [PMID: 25874410 DOI: 10.1002/jhbp.248] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 03/12/2015] [Indexed: 12/12/2022]
Abstract
Heterogeneity is one of the essential hallmarks of malignancies. Within bulk cancer cells, a striking variability differs in biological characteristics including the proliferation rate, cell-cell interaction, metastatic tendency and even sensitivity to anticancer therapies. Such diversity makes the investigation and treatment of the cancers complicated. Increasing evidence suggests this plasticity of cancers is a result of self-renewing and differentiation of a small subpopulation of cancer cells with stem-like properties, called cancer stem cells (CSCs). More importantly, CSCs are believed to be responsible for the resistance to conventional therapies and metastatic abilities in clinical practice. This review summarizes the molecular pathogenesis of hepato-biliary-pancreatic CSCs on the basis of the recent studies, and promising strategy of novel therapy targeting the signal transduction pathways or potentially epigenetic addictions of CSCs.
Collapse
Affiliation(s)
- Shinji Tanaka
- Department of Molecular Oncology, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8519, Japan.
| |
Collapse
|
31
|
Pilapong C, Raiputta C, Chaisupa J, Sittichai S, Thongtem S, Thongtem T. Magnetic-EpCAM nanoprobe as a new platform for efficient targeting, isolating and imaging hepatocellular carcinoma. RSC Adv 2015. [DOI: 10.1039/c5ra01566a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Herein, magnetic-EpCAM nanoparticle (EpCAM-MNP) was developed and exploited as nanoprobe for targeting, isolating and imaging hepatocellular carcinoma.
Collapse
Affiliation(s)
- C. Pilapong
- Center of Excellence for Molecular Imaging (CEMI)
- Department of Radiologic Technology
- Faculty of Associated Medical Sciences
- Chiang Mai University
- Chiang Mai 50200
| | - C. Raiputta
- Center of Excellence for Molecular Imaging (CEMI)
- Department of Radiologic Technology
- Faculty of Associated Medical Sciences
- Chiang Mai University
- Chiang Mai 50200
| | - J. Chaisupa
- Center of Excellence for Molecular Imaging (CEMI)
- Department of Radiologic Technology
- Faculty of Associated Medical Sciences
- Chiang Mai University
- Chiang Mai 50200
| | - S. Sittichai
- Department of Physics and Material Science
- Faculty of Science
- Chiang Mai University
- Chiang Mai 50200
- Thailand
| | - S. Thongtem
- Department of Physics and Material Science
- Faculty of Science
- Chiang Mai University
- Chiang Mai 50200
- Thailand
| | - T. Thongtem
- Department of Chemistry
- Faculty of Science
- Chiang Mai University
- Chiang Mai 50200
- Thailand
| |
Collapse
|
32
|
Gao J, Yan Q, Liu S, Yang X. Knockdown of EpCAM enhances the chemosensitivity of breast cancer cells to 5-fluorouracil by downregulating the antiapoptotic factor Bcl-2. PLoS One 2014; 9:e102590. [PMID: 25019346 PMCID: PMC4097402 DOI: 10.1371/journal.pone.0102590] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/19/2014] [Indexed: 12/19/2022] Open
Abstract
Resistance to fluoropyrimidine-based chemotherapy is the main reason for the failure of cancer treatment, and drug resistance is associated with an inability of tumor cells to undergo apoptosis in response to treatment. Alterations in the expression of epithelial cell adhesion molecule (EpCAM) affect the sensitivity or resistance of tumor cells to anticancer treatment and the activity of intracellular signaling pathways. However, the role of EpCAM in the induction of apoptosis in breast cancer cells remains unclear. Here, we investigated the effect of EpCAM gene knockdown on chemosensitivity to 5-fluorouracil (5-FU) in MCF-7 cells and explored the underlying mechanisms. Our results showed that knockdown of EpCAM promoted apoptosis, inhibited cell proliferation and caused cell-cycle arrest. EpCAM knockdown enhanced the cytotoxic effect of 5-FU, promoting apoptosis by downregulating the expression of the anti-apoptotic protein Bcl-2 and upregulating the expression of the pro-apoptotic proteins Bax, and caspase3 via the ERK1/2 and JNK MAPK signaling pathways in MCF-7 cells. These results indicate that knockdown of EpCAM may have a tumor suppressor effect and suggest EpCAM as a potential target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Jiujiao Gao
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People’s Republic of China
| | - Qiu Yan
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People’s Republic of China
| | - Shuai Liu
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People’s Republic of China
| | - Xuesong Yang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People’s Republic of China
- * E-mail:
| |
Collapse
|