1
|
Liang F, Xu H, Cheng H, Zhao Y, Zhang J. Patient-derived tumor models: a suitable tool for preclinical studies on esophageal cancer. Cancer Gene Ther 2023; 30:1443-1455. [PMID: 37537209 DOI: 10.1038/s41417-023-00652-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/13/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023]
Abstract
Esophageal cancer (EC) is the tenth most common cancer worldwide and has high morbidity and mortality. Its main subtypes include esophageal squamous cell carcinoma and esophageal adenocarcinoma, which are usually diagnosed during their advanced stages. The biological defects and inability of preclinical models to summarize completely the etiology of multiple factors, the complexity of the tumor microenvironment, and the genetic heterogeneity of tumors severely limit the clinical treatment of EC. Patient-derived models of EC not only retain the tissue structure, cell morphology, and differentiation characteristics of the original tumor, they also retain tumor heterogeneity. Therefore, compared with other preclinical models, they can better predict the efficacy of candidate drugs, explore novel biomarkers, combine with clinical trials, and effectively improve patient prognosis. This review discusses the methods and animals used to establish patient-derived models and genetically engineered mouse models, especially patient-derived xenograft models. It also discusses their advantages, applications, and limitations as preclinical experimental research tools to provide an important reference for the precise personalized treatment of EC and improve the prognosis of patients.
Collapse
Affiliation(s)
- Fan Liang
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hongyan Xu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hongwei Cheng
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yabo Zhao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Junhe Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China.
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
2
|
He X, Yan H, Hu J, Duan X, Zhang M, Li H, Wang J, Gao Q, Yu S, Hou X, Liao G, Guo S, Li J, Ge Y, Chen X, Wang W, Tang J. HDS screening with patient-derived primary cells guided individualized therapy for esophageal squamous cell carcinoma- in vivo and vitro. Front Med (Lausanne) 2023; 10:1212851. [PMID: 37601787 PMCID: PMC10433228 DOI: 10.3389/fmed.2023.1212851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/30/2023] [Indexed: 08/22/2023] Open
Abstract
Objective To analyze and evaluate the role of the High-throughput Drug Sensitivity (HDS) screening strategy in identifying highly sensitive drugs against esophageal squamous cell carcinoma (ESCC). Methods A total of 80 patients with progressive ESCC were randomly divided into the observation (40 cases) and the control groups (40 cases). In the observation group, primary ESCC cells were isolated from the tumor tissues with a gastroscope, and drug sensitivity screening was performed on cells derived from the 40 ESCC cases using the HDS method, followed by verification in a patient-derived tumor xenograft (PDX) mouse model. Finally, the differences in the therapeutic efficacy (levels of CEA, CYFRA21-1, SCCA after chemotherapy and the rates of overall survival, local progression, and distant metastasis at 12 months and 18 months time points after chemotherapy) were compared between the observation group (Screened drug-treated) and the control group (Paclitaxel combined with cisplatin regimen-treated). Results Forty ESCC patients were screened for nine different high-sensitive chemotherapeutics, with the majority showing sensitivity to Bortezomib. Experiments on animal models revealed that the tumor tissue mass of PDX mice treated with the HDS-screened drug was significantly lower than that of the Paclitaxel-treated mice (p < 0.05), and the therapeutic efficacy of the observation group was better than the control group (p < 0.05). Conclusion HDS screening technology can be beneficial in screening high-efficacy anticancer drugs for advanced-stage ESCC patients, thereby minimizing adverse drug toxicity in critically ill patients. Moreover, this study provides a new avenue for treating advanced ESCC patients with improved outcomes.
Collapse
Affiliation(s)
- Xing He
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Hezhong Yan
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Jie Hu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Xiaowei Duan
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Mingjin Zhang
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Haiqing Li
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Jiaoxue Wang
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Qian Gao
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Senyuan Yu
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Xilu Hou
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Guobin Liao
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Shicun Guo
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Jin Li
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Yurong Ge
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Xiaolan Chen
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| | - Wenchao Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Jun Tang
- Department of Gastroenterology, The 901th Hospital of Joint Logistics Support Force, Hefei, Anhui, China
| |
Collapse
|
3
|
Liu H, Wang X. Esophageal organoids: applications and future prospects. J Mol Med (Berl) 2023; 101:931-945. [PMID: 37380866 DOI: 10.1007/s00109-023-02340-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/26/2023] [Accepted: 06/14/2023] [Indexed: 06/30/2023]
Abstract
Organoids have been developed in the last decade as a new research tool to simulate organ cell biology and disease. Compared to traditional 2D cell lines and animal models, experimental data based on esophageal organoids are more reliable. In recent years, esophageal organoids derived from multiple cell sources have been established, and relatively mature culture protocols have been developed. Esophageal inflammation and cancer are two directions of esophageal organoid modeling, and organoid models of esophageal adenocarcinoma, esophageal squamous cell carcinoma, and eosinophilic esophagitis have been established. The properties of esophageal organoids, which mimic the real esophagus, contribute to research in drug screening and regenerative medicine. The combination of organoids with other technologies, such as organ chips and xenografts, can complement the deficiencies of organoids and create entirely new research models that are more advantageous for cancer research. In this review, we will summarize the development of tumor and non-tumor esophageal organoids, the current application of esophageal organoids in disease modeling, regenerative medicine, and drug screening. We will also discuss the future prospects of esophageal organoids.
Collapse
Affiliation(s)
- Hongyuan Liu
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xianli Wang
- Shanghai Jiao Tong University, School of Public Health, Shanghai, 200025, China.
| |
Collapse
|
4
|
Li Q, Tie Y, Alu A, Ma X, Shi H. Targeted therapy for head and neck cancer: signaling pathways and clinical studies. Signal Transduct Target Ther 2023; 8:31. [PMID: 36646686 PMCID: PMC9842704 DOI: 10.1038/s41392-022-01297-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/27/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023] Open
Abstract
Head and neck cancer (HNC) is malignant, genetically complex and difficult to treat and is the sixth most frequent cancer, with tobacco, alcohol and human papillomavirus being major risk factors. Based on epigenetic data, HNC is remarkably heterogeneous, and treatment remains challenging. There is a lack of significant improvement in survival and quality of life in patients with HNC. Over half of HNC patients experience locoregional recurrence or distal metastasis despite the current multiple traditional therapeutic strategies and immunotherapy. In addition, resistance to chemotherapy, radiotherapy and some targeted therapies is common. Therefore, it is urgent to explore more effective and tolerable targeted therapies to improve the clinical outcomes of HNC patients. Recent targeted therapy studies have focused on identifying promising biomarkers and developing more effective targeted therapies. A well understanding of the pathogenesis of HNC contributes to learning more about its inner association, which provides novel insight into the development of small molecule inhibitors. In this review, we summarized the vital signaling pathways and discussed the current potential therapeutic targets against critical molecules in HNC, as well as presenting preclinical animal models and ongoing or completed clinical studies about targeted therapy, which may contribute to a more favorable prognosis of HNC. Targeted therapy in combination with other therapies and its limitations were also discussed.
Collapse
Affiliation(s)
- Qingfang Li
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Tie
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Aqu Alu
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Wang Z, Cormier RT. Golden Syrian Hamster Models for Cancer Research. Cells 2022; 11:2395. [PMID: 35954238 PMCID: PMC9368453 DOI: 10.3390/cells11152395] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022] Open
Abstract
The golden Syrian hamster (Mesocricetus auratus) has long been a valuable rodent model of human diseases, especially infectious and metabolic diseases. Hamsters have also been valuable models of several chemically induced cancers such as the DMBA-induced oral cheek pouch cancer model. Recently, with the application of CRISPR/Cas9 genetic engineering technology, hamsters can now be gene targeted as readily as mouse models. This review describes the phenotypes of three gene-targeted knockout (KO) hamster cancer models, TP53, KCNQ1, and IL2RG. Notably, these hamster models demonstrate cancer phenotypes not observed in mouse KOs. In some cases, the cancers that arise in the KO hamster are similar to cancers that arise in humans, in contrast with KO mice that do not develop the cancers. An example is the development of aggressive acute myelogenous leukemia (AML) in TP53 KO hamsters. The review also presents a discussion of the relative strengths and weaknesses of mouse cancer models and hamster cancer models and argues that there are no perfect rodent models of cancer and that the genetically engineered hamster cancer models can complement mouse models and expand the suite of animal cancer models available for the development of new cancer therapies.
Collapse
Affiliation(s)
- Zhongde Wang
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Robert T. Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| |
Collapse
|
6
|
Huang Z, Wang C, Zhao X. circFIG 4 drives the carcinogenesis and metastasis of esophagus cancer via the miR-493-5p/E2F3 axis. Thorac Cancer 2022; 13:783-794. [PMID: 35083866 PMCID: PMC8930455 DOI: 10.1111/1759-7714.14321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 01/02/2023] Open
Abstract
Background Esophageal cancer (EC) is a highly malignant tumor of the digestive tract. Circular RNAs (circRNAs) have been verified to play a regulatory role in the occurrence and progression of different cancers, including EC. This research aimed to investigate the role and molecular mechanism of circFIG 4 in EC progression. Methods The analyses of circFIG 4, miR‐493‐5p, and neuro‐oncological ventral antigen 2 levels were administrated by quantitative real‐time polymerase chain reaction. The characteristics of circFIG 4 were determined by Ribonuclease R assay and Actinomycin D assay. Cell proliferation was assessed via colony formation assay and 5‐ethynyl‐2′‐deoxyuridine incorporation assay. Cell cycle distribution and apoptosis were evaluated by flow cytometry. Western blot was performed to assess protein expression. The targeted interaction among circFIG 4, miR‐493‐5p, and E2F transcription factor 3 (E2F3) were validated using dual‐luciferase reporter or RNA immunoprecipitation assays. Results circFIG 4 was overtly upregulated in EC and was relatively stable in EC cells. circFIG 4 knockdown impeded proliferation, migration, and invasion and expedited apoptosis in EC cells. circFIG 4 served as a miR‐493‐5p sponge to act in the development of EC. Furthermore, circFIG 4 modulated EC progression via targeting miR‐493‐5p and miR‐493‐5p suppressed EC progression via targeting E2F3. circFIG 4 modulated E2F3 expression through acting as a sponge of miR‐493‐5p. Moreover, circFIG 4 knockdown inhibited EC tumorigenesis by targeting miR‐493‐5p/E2F3 axis tumor growth in vivo. Conclusion circFIG 4 silence mitigated EC malignant progression at least partly by mediating the miR‐493‐5p/E2F3 pathway, highlighting new biomarkers and therapeutic targets for EC treatment.
Collapse
Affiliation(s)
- Zhen Huang
- Department of Thoracic Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Chunyue Wang
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Fujian Medical University, Xiamen, China
| | - Xin Zhao
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Fujian Medical University, Xiamen, China
| |
Collapse
|