1
|
Huang C, Li Y, Zhang F, Zhang C, Ding Z. Advancements in elucidating the mechanisms of Sorafenib resistance in hepatocellular carcinoma. Int J Surg 2025; 111:2990-3005. [PMID: 39992113 DOI: 10.1097/js9.0000000000002294] [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: 11/14/2024] [Accepted: 01/17/2025] [Indexed: 02/25/2025]
Abstract
Primary liver cancer is a major global health challenge, of which hepatocellular carcinoma is the most common. For patients with advanced liver cancer, Sorafenib is a first-line targeted drug that occupies a dominant position in clinical applications. Sorafenib is a multi-kinase inhibitor commonly used in clinical practice, which can effectively inhibit tumor cell proliferation, promote cell apoptosis, and inhibit angiogenesis. However, the emergence of drug resistance has hindered the development of treatment programs, which is an urgent problem to be solved. Recent studies have revealed many mechanisms and influencing factors of Sorafenib resistance (such as epigenetic regulation, programmed cell death, metabolic reprogramming, and tumor microenvironment changes). This review not only summarizes the above mechanisms, but also summarizes the combined application of Sorafenib with other drugs (such as molecular targeted drugs, other anti-angiogenesis drugs, cytotoxic drugs, immunotherapy drugs, etc .). Finally, potential strategies and research directions to overcome drug resistance (such as targeting epigenetic pathways or metabolic reprogramming) are discussed to provide suggestions for future in-depth research and clinical applications.
Collapse
Affiliation(s)
- Chen Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yangqian Li
- Frontiers Science Center for Disease-related Molecular Network, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fengmei Zhang
- Frontiers Science Center for Disease-related Molecular Network, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chenliang Zhang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenyu Ding
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Yuan J, Yang L, Zhang H, Beeraka NM, Zhang D, Wang Q, Wang M, Pr HV, Sethi G, Wang G. Decoding tumor microenvironment: EMT modulation in breast cancer metastasis and therapeutic resistance, and implications of novel immune checkpoint blockers. Biomed Pharmacother 2024; 181:117714. [PMID: 39615165 DOI: 10.1016/j.biopha.2024.117714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Tumor microenvironment (TME) and epithelial-mesenchymal transition (EMT) play crucial roles in the initiation and progression of tumors. TME is composed of various cell types, such as immune cells, fibroblasts, and endothelial cells, as well as non-cellular components like extracellular matrix (ECM) proteins and soluble factors. These elements interact with tumor cells through a complex network of signaling pathways involving cytokines, growth factors, metabolites, and non-coding RNA-carrying exosomes. Hypoxic conditions within the TME further modulate these interactions, collectively influencing tumor growth, metastatic potential, and response to therapy. EMT represents a dynamic and reversible process where epithelial cells undergo phenotypic changes to adopt mesenchymal characteristics in several cancers, including breast cancers. This transformation enhances cell motility and imparts stem cell-like properties, which are closely associated with increased metastatic capability and resistance to conventional cancer treatments. Thus, understanding the crosstalk between the TME and EMT is essential for unraveling the underlying mechanisms of breast cancer metastasis and therapeutic resistance. This review uniquely examines the intricate interplay between the tumor TME and epithelial-mesenchymal transition EMT in driving breast cancer metastasis and treatment resistance. It explores the therapeutic potential of targeting the TME-EMT axis, specifically through CD73-TGF-β dual-blockade, to improve outcomes in triple-negative breast cancer. Additionally, it underscores new strategies to enhance immune checkpoint blockade (ICB) responses by modulating EMT, thereby offering innovative insights for more effective cancer treatment.
Collapse
Affiliation(s)
- Jie Yuan
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Li Yang
- Department of Clinical Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Hua Zhang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Narasimha M Beeraka
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia; Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Chiyyedu, Anantapuramu, Andhra Pradesh 515721, India; Department of Studies in Molecular Biology, Faculty of Science and Technology, University of Mysore, Mysore, Karnataka, 570006, India.
| | - Danfeng Zhang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Qun Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Minghua Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Hemanth Vikram Pr
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Geng Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| |
Collapse
|
3
|
Ye G, Ye M, Jin X. Roles of clinical application of lenvatinib and its resistance mechanism in advanced hepatocellular carcinoma (Review). Am J Cancer Res 2024; 14:4113-4171. [PMID: 39417171 PMCID: PMC11477829 DOI: 10.62347/ujvp4361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Lenvatinib (LEN) is a multi-target TKI, which plays a pivotal role in the treatment of advanced hepatocellular carcinoma (HCC). The inevitable occurrence of drug resistance still prevents curative potential and is deleterious for the prognosis, and a growing body of studies is accumulating, which have devoted themselves to unveiling its underlying resistance mechanism and made some progress. The dysregulation of crucial signaling pathways, non-coding RNA and RNA modifications were proven to be associated with LEN resistance. A range of drugs were found to influence LEN therapeutic efficacy. In addition, the superiority of LEN combination therapy has been shown to potentially overcome the limitations of LEN monotherapy in a series of research, and a range of promising indicators for predicting treatment response and prognosis have been discovered in recent years. In this review, we summarize the latest developments in LEN resistance, the efficacy and safety of LEN combination therapy as well as associated indicators, which may provide new insight into its resistance as well as ideas in the treatment of advanced HCC.
Collapse
Affiliation(s)
- Ganghui Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
- Department of Radiation Oncology, Taizhou Central Hospital (Taizhou University Hospital)Taizhou 318000, Zhejiang, P. R. China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
| |
Collapse
|
4
|
Wang L, Tong L, Xiong Z, Chen Y, Zhang P, Gao Y, Liu J, Yang L, Huang C, Ye G, Du J, Liu H, Yang W, Wang Y. Ferroptosis-inducing nanomedicine and targeted short peptide for synergistic treatment of hepatocellular carcinoma. J Nanobiotechnology 2024; 22:533. [PMID: 39223666 PMCID: PMC11370132 DOI: 10.1186/s12951-024-02808-7] [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: 02/19/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The poor prognosis of hepatocellular carcinoma (HCC) is still an urgent challenge to be solved worldwide. Hence, assembling drugs and targeted short peptides together to construct a novel medicine delivery strategy is crucial for targeted and synergy therapy of HCC. Herein, a high-efficiency nanomedicine delivery strategy has been constructed by combining graphdiyne oxide (GDYO) as a drug-loaded platform, specific peptide (SP94-PEG) as a spear to target HCC cells, sorafenib, doxorubicin-Fe2+ (DOX-Fe2+), and siRNA (SLC7A11-i) as weapons to exert a three-path synergistic attack against HCC cells. In this work, SP94-PEG and GDYO form nanosheets with HCC-targeting properties, the chemotherapeutic drug DOX linked to ferrous ions increases the free iron pool in HCC cells and synergizes with sorafenib to induce cell ferroptosis. As a key gene of ferroptosis, interference with the expression of SLC7A11 makes the ferroptosis effect in HCC cells easier, stronger, and more durable. Through gene interference, drug synergy, and short peptide targeting, the toxic side effects of chemotherapy drugs are reduced. The multifunctional nanomedicine GDYO@SP94/DOX-Fe2+/sorafenib/SLC7A11-i (MNMG) possesses the advantages of strong targeting, good stability, the ability to continuously induce tumor cell ferroptosis and has potential clinical application value, which is different from traditional drugs.
Collapse
Affiliation(s)
- Luyang Wang
- Department of Clinical Research Center, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, P. R. China
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
- Center for Drug Safety Evaluation, Qingdao Center for Pharmaceutical Collaborative Innovation, Qingdao, 266209, Shandong, P. R. China
- Department of Laboratory Medicine, Xinhua Hospital of Zhejiang Province, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China
| | - Le Tong
- Department of Clinical Research Center, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, P. R. China
- Department of Laboratory Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, P. R. China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, P. R. China
| | - Zecheng Xiong
- CAS Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, CAS Research/ Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yi Chen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Ping Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Yan Gao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Jing Liu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Lei Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Chunqi Huang
- Department of Laboratory Medicine, Xinhua Hospital of Zhejiang Province, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China
| | - Gaoqi Ye
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China.
| | - Huibiao Liu
- CAS Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, CAS Research/ Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, P. R. China.
| | - Wei Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China.
| | - Ying Wang
- Department of Clinical Research Center, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, P. R. China.
- Department of Laboratory Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, P. R. China.
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China.
| |
Collapse
|
5
|
Yao Y, Zhao Q, Xu F, Yao T. Enhanced anti-tumor therapy for hepatocellular carcinoma via sorafenib and KIAA1199-siRNA co-delivery liposomes. Saudi Pharm J 2024; 32:102153. [PMID: 39211513 PMCID: PMC11357851 DOI: 10.1016/j.jsps.2024.102153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignancies worldwide. Sorafenib (Sf) is currently the first-line treatment for HCC. However, due to the side effects and unsatisfied efficiency of Sf, it is urgent to combine different therapeutic agents to inhibit HCC progression and increase the therapeutic efficacy. Here, our study constructed a Sf and KIAA1199-siRNA co-loaded liposome Sf-Lp-KIAA, which was prepared by electrostatic interaction of KIAA1199-siRNA and Sf loaded liposome (Sf-Lp). The particle size, zeta potential, the in vitro cumulative release was investigated. The physical and chemical properties were characterized, and the inhibition of HepG2 growth and metastasis in vitro was investigated. The cellular uptake of the co-loaded liposome was significantly higher than that of free siRNA, and the drug/siRNA could be co-delivered to the target cells. Sf-Lp-KIAA could significantly inhibit the growth, migration, invasion and down-regulate KIAA1199 expression of HepG2 cells in vitro than that of single Sf treated group. In addition, the co-delivery liposome accumulated in the HepG2 subcutaneous tumor model and suppress tumor growth after systemic administration without induce obvious toxicity. The present study implied that the co-delivery of Sf and KIAA1199-siRNA through the co-loaded liposomes exerted synergistic antitumor effects on HCC, which would lay a foundation for HCC therapy in the future.
Collapse
Affiliation(s)
- Yao Yao
- Guangdong Food and Drug Vocational College, Guangzhou, China
| | - Qian Zhao
- Department of Gynecological Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Feng Xu
- Guangdong Food and Drug Vocational College, Guangzhou, China
| | - Tingting Yao
- Department of Gynecological Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
6
|
Heydarnia E, Sepasi A, Asefi N, Khakshournia S, Mohammadnejad J. The effects of metformin and PCL-sorafenib nanoparticle co-treatment on MCF-7 cell culture model of breast cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7213-7221. [PMID: 38656346 DOI: 10.1007/s00210-024-03049-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 03/10/2024] [Indexed: 04/26/2024]
Abstract
Despite breakthrough therapeutics in breast cancer, it is one of the main causes of mortality among women worldwide. Thus, drug therapies for treating breast cancer have recently been developed by scientists. Metformin and sorafenib are well-known therapeutics in breast cancer. In the present study, we combined sorafenib and PCL-sorafenib with metformin to improve drug absorption and promote therapeutic efficiency. The MCF-7 cells were treated with metformin, sorafenib, or PCL-sorafenib. The growth inhibitory effect of these drugs and cell viability were assessed using MTT and flow cytometry assays, respectively. The expression of targeted genes involved in cell proliferation, signaling, and the cell cycle was measured by real-time PCR. The results showed that MCF-7 cells treated with metformin/sorafenib and PCL-sorafenib/metformin co-treatment contributed to 50% viability compared to the untreated group. Moreover, PI and Annexin V staining tests showed that the cell viability for metformin/sorafenib and PCL-sorafenib/metformin was 38% and 17%, respectively. Furthermore, sorafenib/metformin and PCL-sorafenib/metformin lead to p53 gene expression increase by which they can increase ROS, thereby decreasing GPX4 gene expression. In addition, they affected the expression of BCL2 and BAX genes and altered the cell cycle. Together, the combination of PCL-sorafenib/metformin and sorafenib/metformin increased sorafenib absorption at lower doses and also led to apoptosis and oxidative stress increases in MCF-7 cells.
Collapse
Affiliation(s)
- Emad Heydarnia
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Aref Sepasi
- Department of Medical Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Genetics, Breast Cancer Research Center, Motamed Cancer Institute, Tehran, Iran
| | - Nika Asefi
- Department of Genetics, Breast Cancer Research Center, Motamed Cancer Institute, Tehran, Iran
| | - Sara Khakshournia
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Javad Mohammadnejad
- Department of Life Science Engineering, Faculty of New Science and Technologies, University of Tehran, Tehran, 14395-1561, Iran.
| |
Collapse
|
7
|
Qiu P, Zhou K, Wang Y, Chen X, Xiao C, Li W, Chen Y, Chang Y, Liu J, Zhou F, Wang X, Shang J, Liu L, Qiu Z. Revitalizing gut barrier integrity: role of miR-192-5p in enhancing autophagy via Rictor in enteritis. Am J Physiol Gastrointest Liver Physiol 2024; 327:G317-G332. [PMID: 38954822 DOI: 10.1152/ajpgi.00291.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
Intestinal inflammation and compromised barrier function are critical factors in the pathogenesis of gastrointestinal disorders. This study aimed to investigate the role of miR-192-5p in modulating intestinal epithelial barrier (IEB) integrity and its association with autophagy. A DSS-induced colitis model was used to assess the effects of miR-192-5p on intestinal inflammation. In vitro experiments involved cell culture and transient transfection techniques. Various assays, including dual-luciferase reporter gene assays, quantitative real-time PCR, Western blotting, and measurements of transepithelial electrical resistance, were performed to evaluate changes in miR-192-5p expression, Rictor levels, and autophagy flux. Immunofluorescence staining, H&E staining, TEER measurements, and FITC-dextran analysis were also used. Our findings revealed a reduced expression of miR-192-5p in inflamed intestinal tissues, correlating with impaired IEB function. Overexpression of miR-192-5p alleviated TNF-induced IEB dysfunction by targeting Rictor, resulting in enhanced autophagy flux in enterocytes (ECs). Moreover, the therapeutic potential of miR-192-5p was substantiated in colitis mice, wherein increased miR-192-5p expression ameliorated intestinal inflammatory injury by enhancing autophagy flux in ECs through the modulation of Rictor. Our study highlights the therapeutic potential of miR-192-5p in enteritis by demonstrating its role in regulating autophagy and preserving IEB function. Targeting the miR-192-5p/Rictor axis is a promising approach for mitigating gut inflammatory injury and improving barrier integrity in patients with enteritis.NEW & NOTEWORTHY We uncover the pivotal role of miR-192-5p in fortifying intestinal barriers amidst inflammation. Reduced miR-192-5p levels correlated with compromised gut integrity during inflammation. Notably, boosting miR-192-5p reversed gut damage by enhancing autophagy via suppressing Rictor, offering a potential therapeutic strategy for fortifying the intestinal barrier and alleviating inflammation in patients with enteritis.
Collapse
Affiliation(s)
- Peishan Qiu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Kezhi Zhou
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Youwei Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Xiaoyu Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Cong Xiao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Wenjie Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Yuhua Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Ying Chang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Jing Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Feng Zhou
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Xiaobing Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Jian Shang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Lan Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Zhao Qiu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| |
Collapse
|
8
|
Liu Y, Shao K, Yang W, Shen Q, Lu M, Shao Z, Chu S, Wang Y, Wang X, Chen X, Bai J, Wu X. Phosphorylated FOXQ1, a novel substrate of JNK1, inhibits sorafenib-induced ferroptosis by activating ETHE1 in hepatocellular carcinoma. Cell Death Dis 2024; 15:395. [PMID: 38839744 PMCID: PMC11153576 DOI: 10.1038/s41419-024-06789-1] [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: 07/05/2023] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous and malignant cancer with poor overall survival. The application of sorafenib is a major breakthrough in the treatment of HCC. In our study, FOXQ1 was significantly overexpressed in sorafenib-resistant HCC cells and suppressed sorafenib-induced ferroptosis. We found that phosphorylation of FOXQ1 at serine 248 is critical for the suppression of sorafenib-induced ferroptosis. Furthermore, as the upstream phosphorylation kinase of FOXQ1, JNK1, which is activated by sorafenib, can directly phosphorylate the serine 248 site of FOXQ1. Then, the phosphorylated FOXQ1 got a high affinity for the promoter of ETHE1 and activates its transcription. Further flow cytometry results showed that ETHE1 reduced intracellular lipid peroxidation and iron levels. Collectively, our study implicated the JNK1-FOXQ1-ETHE1 axis in HCC ferroptosis induced by sorafenib, providing mechanistic insight into sensitivity to sorafenib therapy of HCC.
Collapse
Affiliation(s)
- Yiwei Liu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Ke Shao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
- Department of General Surgery, The People's Hospital of Rugao, Affiliated Rugao Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Wendong Yang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Qi Shen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Mengru Lu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Zhiying Shao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Sufang Chu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yuming Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China.
| | - Xiaofeng Chen
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Xiaofeng Wu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China.
| |
Collapse
|
9
|
Hu Z, Zhao Y, Li L, Jiang J, Li W, Mang Y, Gao Y, Dong Y, Zhu J, Yang C, Ran J, Li L, Zhang S. Metformin promotes ferroptosis and sensitivity to sorafenib in hepatocellular carcinoma cells via ATF4/STAT3. Mol Biol Rep 2023; 50:6399-6413. [PMID: 37326750 PMCID: PMC10374833 DOI: 10.1007/s11033-023-08492-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/28/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common cancer worldwide, and sorafenib is a first-line drug for the treatment of advanced liver cancer. Resistance to sorafenib has become a major challenge in the treatment of hepatocellular carcinoma, however, studies have shown that metformin can promote ferroptosis and sorafenib sensitivity. Therefore, the aim of this study was to investigate the promotion of ferroptosis and sorafenib sensitivity by metformin via ATF4/STAT3 in hepatocellular carcinoma cells. METHODS Hepatocellular carcinoma cells Huh7 and Hep3B and induced sorafenib resistance (SR) Huh7/SR and Hep3B/SR cells were used as in vitro cell models. Cells were injected subcutaneously to establish a drug-resistant mouse model. CCK-8 was used to detect cell viability and sorafenib IC50. Western blotting was used to detect the expression of relevant proteins. BODIPY staining was used to analyze the lipid peroxidation level in cells. A scratch assay was used to detect cell migration. Transwell assays were used to detect cell invasion. Immunofluorescence was used to localize the expression of ATF4 and STAT3. RESULTS Metformin promoted ferroptosis in hepatocellular carcinoma cells through ATF4/STAT3, decreased sorafenib IC50, increased ROS and lipid peroxidation levels, decreased cell migration and invasion, inhibited the expression of the drug-resistant proteins ABCG2 and P-GP in hepatocellular carcinoma cells, and thus inhibited sorafenib resistance in hepatocellular carcinoma cells. Downregulating ATF4 inhibited the phosphorylated nuclear translocation of STAT3, promoted ferroptosis, and increased the sensitivity of Huh7 cells to sorafenib. Metformin was also shown in animal models to promote ferroptosis and sorafenib sensitivity in vivo via ATF4/STAT3. CONCLUSION Metformin promotes ferroptosis and sensitivity to sorafenib in hepatocellular carcinoma cells via ATF4/STAT3, and it inhibits HCC progression.
Collapse
Affiliation(s)
- Zongqiang Hu
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Yingpeng Zhao
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Laibang Li
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jie Jiang
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Wang Li
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Yuanyi Mang
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Yang Gao
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Yun Dong
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jiashun Zhu
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Chaomin Yang
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jianghua Ran
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China.
| | - Li Li
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China.
| | - Shengning Zhang
- Hepato-pancreato-biliary Surgery Department, First People's Hospital of Kunming City & The Calmette Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China.
| |
Collapse
|
10
|
You AB, Yang H, Lai CP, Lei W, Yang L, Lin JL, Liu SC, Ding N, Ye F. CMTR1 promotes colorectal cancer cell growth and immune evasion by transcriptionally regulating STAT3. Cell Death Dis 2023; 14:245. [PMID: 37024465 PMCID: PMC10079662 DOI: 10.1038/s41419-023-05767-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 04/08/2023]
Abstract
CMTR1, also called IFN-stimulated gene 95 kDa protein (ISG95), is elevated by viral infection in a variety of cells. However, the functions of CMTR1 in colorectal cancer (CRC), especially its roles in tumorigenesis and immune regulation, remain unclear. Here, we first identified CMTR1 as a novel oncogene in colorectal cancer. Based on The Cancer Genome Atlas (TCGA) database exploration and human tissue microarray (TMA) analysis, we found that CMTR1 expression was markedly higher in CRC tissues than in adjacent normal tissues. High CMTR1 expression was correlated with poor prognosis in CRC patients. Knockdown (KD) of CMTR1 significantly suppressed cell proliferation and tumorigenicity both in vitro and in vivo, whereas overexpression of CMTR1 resulted in the opposite effects. KEGG pathway analysis revealed differential enrichment in the JAK/STAT signaling pathway in colorectal cancer cells with CMTR1 KD. Mechanistically, suppression of CMTR1 expression inhibited RNAPII recruitment to the transcription start site (TSS) of STAT3 and suppressed STAT3 expression and activation. Furthermore, the efficacy of PD1 blockade immunotherapy was prominently enhanced in the presence of CMTR1 KD via increased infiltration of CD8 + T cells into the tumor microenvironment. Overall, it appears that CMTR1 plays a key role in regulating tumor cell proliferation and antitumor immunity.
Collapse
Affiliation(s)
- A-Bin You
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, 350122, China
| | - Hu Yang
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, 350122, China
| | - Chun-Ping Lai
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, 350122, China
| | - Wen Lei
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, 350122, China
| | - Lu Yang
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, 350122, China
| | - Jia-Lin Lin
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, 350122, China
| | - Shun-Cui Liu
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, 350122, China.
- Department of Anesthesiology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China.
| | - Nan Ding
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China.
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, 350122, China.
| | - Feng Ye
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China.
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, 350122, China.
| |
Collapse
|
11
|
Wang L, Xu X, Chen D, Li C. Dihydrotanshinone I inhibits hepatocellular carcinoma cells proliferation through DNA damage and EGFR pathway. PeerJ 2023; 11:e15022. [PMID: 36935927 PMCID: PMC10019332 DOI: 10.7717/peerj.15022] [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: 09/19/2022] [Accepted: 02/17/2023] [Indexed: 03/16/2023] Open
Abstract
Background The incidence and mortality of hepatocellular carcinoma (HCC) are globally on the rise. Dihydrotanshinone I, a natural product isolated from Salvia miltiorrhiza Bunge, has attracted extensive attention in recent years for its anti-tumour proliferation efficiency. Methods Cell proliferations in hepatoma cells (Huh-7 and HepG2) were evaluated by MTT and colony formation assays. Immunofluorescence (IF) of 53BP1 and flow cytometry analysis were performed to detect DNA damage and cell apoptosis. Furthermore, network pharmacological analysis was applied to explore the potential therapeutic targets and pathway of dihydrotanshinone I. Results The results showed that dihydrotanshinone I effectively inhibited the proliferation of Huh-7 and HepG2 cells. Moreover, dihydrotanshinone I dose-dependently induced DNA-damage and apoptosis in vitro. Network pharmacological analysis and molecular simulation results indicated that EGFR might be a potential therapeutic target of dihydrotanshinone I in HCC. Collectively, our findings suggested that dihydrotanshinone I is a novel candidate therapeutic agent for HCC treatment.
Collapse
Affiliation(s)
- Linjun Wang
- Department of Hepatopancreatobiliary Surgery, The First People’s Hospital of Yongkang, Yongkang, Zhejiang, China
| | - Xiangwei Xu
- Department of Pharmacy, The First People’s Hosipital of Yongkang, Yongkang, Zhejiang, China
| | - Dexing Chen
- Department of Hepatopancreatobiliary Surgery, The First People’s Hospital of Yongkang, Yongkang, Zhejiang, China
| | - Chenghang Li
- Department of Infectious Liver Disease, The First People’s Hospital of Yongkang, Yongkang, Zhejiang, China
| |
Collapse
|
12
|
Yan ZJ, Chen L, Wang HY. To be or not to be: The double-edged sword roles of liver progenitor cells. Biochim Biophys Acta Rev Cancer 2023; 1878:188870. [PMID: 36842766 DOI: 10.1016/j.bbcan.2023.188870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/11/2023] [Accepted: 01/28/2023] [Indexed: 02/28/2023]
Abstract
Given the liver's remarkable and unique regenerative capacity, researchers have long focused on liver progenitor cells (LPCs) and liver cancer stem cells (LCSCs). LPCs can differentiate into both hepatocytes and cholangiocytes. However, the mechanism underlying cell conversion and its distinct contribution to liver homeostasis and tumorigenesis remain unclear. In this review, we discuss the complicated conversions involving LPCs and LCSCs. As the critical intermediate state in malignant transformation, LPCs play double-edged sword roles. LPCs are not only involved in hepatic wound-healing responses by supplementing liver cells and bile duct cells in the damaged liver but may transform into LCSCs under dysregulation of key signaling pathways, resulting in refractory malignant liver tumors. Because LPC lineages are temporally and spatially dynamic, we discuss crucial LPC subgroups and summarize regulatory factors correlating with the trajectories of LPCs and LCSCs in the liver tumor microenvironment. This review elaborates on the double-edged sword roles of LPCs to help understand the liver's regenerative potential and tumor heterogeneity. Understanding the sources and transformations of LPCs is essential in determining how to exploit their regenerative capacity in the future.
Collapse
Affiliation(s)
- Zi-Jun Yan
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Shanghai 200438, PR China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, PR China; Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, PR China
| | - Lei Chen
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Shanghai 200438, PR China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, PR China; Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, PR China.
| | - Hong-Yang Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Shanghai 200438, PR China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education, Shanghai 200438, PR China; Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai 200438, PR China.
| |
Collapse
|
13
|
Harnessing epithelial-mesenchymal plasticity to boost cancer immunotherapy. Cell Mol Immunol 2023; 20:318-340. [PMID: 36823234 PMCID: PMC10066239 DOI: 10.1038/s41423-023-00980-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/17/2023] [Indexed: 02/25/2023] Open
Abstract
Immune checkpoint blockade (ICB) therapy is a powerful option for cancer treatment. Despite demonstrable progress, most patients fail to respond or achieve durable responses due to primary or acquired ICB resistance. Recently, tumor epithelial-to-mesenchymal plasticity (EMP) was identified as a critical determinant in regulating immune escape and immunotherapy resistance in cancer. In this review, we summarize the emerging role of tumor EMP in ICB resistance and the tumor-intrinsic or extrinsic mechanisms by which tumors exploit EMP to achieve immunosuppression and immune escape. We discuss strategies to modulate tumor EMP to alleviate immune resistance and to enhance the efficiency of ICB therapy. Our discussion provides new prospects to enhance the ICB response for therapeutic gain in cancer patients.
Collapse
|
14
|
Jiang Z, Dai C. Potential Treatment Strategies for Hepatocellular Carcinoma Cell Sensitization to Sorafenib. J Hepatocell Carcinoma 2023; 10:257-266. [PMID: 36815094 PMCID: PMC9939808 DOI: 10.2147/jhc.s396231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Liver cancer is highly malignant, has a low sensitivity to chemotherapy, and is associated with poor patient prognosis. The last 3 years have seen the emergence of promising targeted therapies for the treatment of hepatocellular carcinoma (HCC). For over 10 years, before the discovery of lenvatinib, sorafenib was only first-line therapeutic agent available for the treatment of advanced HCC. However, several clinical studies have shown that a considerable proportion liver cancer patients are insensitive to sorafenib. Very few patients actually substantially benefit from treatment with sorafenib, and the overall efficacy of the drug has not been satisfactory; therefore, sorafenib has attracted considerable research attention. This study, which is based on previous studies and reports, reviews the potential mechanisms underlying sorafenib resistance and summarizes combination therapies and potential drugs that can be used to sensitize HCC cells to sorafenib.
Collapse
Affiliation(s)
- Zhonghao Jiang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Chaoliu Dai
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China,Correspondence: Chaoliu Dai, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China, Email
| |
Collapse
|
15
|
Tian X, Yan T, Liu F, Liu Q, Zhao J, Xiong H, Jiang S. Link of sorafenib resistance with the tumor microenvironment in hepatocellular carcinoma: Mechanistic insights. Front Pharmacol 2022; 13:991052. [PMID: 36071839 PMCID: PMC9441942 DOI: 10.3389/fphar.2022.991052] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 07/25/2022] [Indexed: 11/26/2022] Open
Abstract
Sorafenib, a multi-kinase inhibitor with antiangiogenic, antiproliferative, and proapoptotic properties, is the first-line treatment for patients with late-stage hepatocellular carcinoma (HCC). However, the therapeutic effect remains limited due to sorafenib resistance. Only about 30% of HCC patients respond well to the treatment, and the resistance almost inevitably happens within 6 months. Thus, it is critical to elucidate the underlying mechanisms and identify effective approaches to improve the therapeutic outcome. According to recent studies, tumor microenvironment (TME) and immune escape play critical roles in tumor occurrence, metastasis and anti-cancer drug resistance. The relevant mechanisms were focusing on hypoxia, tumor-associated immune-suppressive cells, and immunosuppressive molecules. In this review, we focus on sorafenib resistance and its relationship with liver cancer immune microenvironment, highlighting the importance of breaking sorafenib resistance in HCC.
Collapse
Affiliation(s)
- Xinchen Tian
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tinghao Yan
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fen Liu
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Qingbin Liu
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Jing Zhao
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, China
- *Correspondence: Huabao Xiong, ; Shulong Jiang,
| | - Shulong Jiang
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
- *Correspondence: Huabao Xiong, ; Shulong Jiang,
| |
Collapse
|
16
|
Huang M, Lin Y, Wang C, Deng L, Chen M, Assaraf YG, Chen ZS, Ye W, Zhang D. New insights into antiangiogenic therapy resistance in cancer: Mechanisms and therapeutic aspects. Drug Resist Updat 2022; 64:100849. [PMID: 35842983 DOI: 10.1016/j.drup.2022.100849] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Angiogenesis is a hallmark of cancer and is required for tumor growth and progression. Antiangiogenic therapy has been revolutionarily developing and was approved for the treatment of various types of cancer for nearly two decades, among which bevacizumab and sorafenib continue to be the two most frequently used antiangiogenic drugs. Although antiangiogenic therapy has brought substantial survival benefits to many cancer patients, resistance to antiangiogenic drugs frequently occurs during clinical treatment, leading to poor outcomes and treatment failure. Cumulative evidence has demonstrated that the intricate interplay among tumor cells, bone marrow-derived cells, and local stromal cells critically allows for tumor escape from antiangiogenic therapy. Currently, drug resistance has become the main challenge that hinders the therapeutic efficacies of antiangiogenic therapy. In this review, we describe and summarize the cellular and molecular mechanisms conferring tumor drug resistance to antiangiogenic therapy, which was predominantly associated with redundancy in angiogenic signaling molecules (e.g., VEGFs, GM-CSF, G-CSF, and IL17), alterations in biological processes of tumor cells (e.g., tumor invasiveness and metastasis, stemness, autophagy, metabolic reprogramming, vessel co-option, and vasculogenic mimicry), increased recruitment of bone marrow-derived cells (e.g., myeloid-derived suppressive cells, tumor-associated macrophages, and tumor-associated neutrophils), and changes in the biological functions and features of local stromal cells (e.g., pericytes, cancer-associated fibroblasts, and endothelial cells). We also review potential biomarkers to predict the response to antiangiogenic therapy in cancer patients, which mainly consist of imaging biomarkers, cellular and extracellular proteins, a certain type of bone marrow-derived cells, local stromal cell content (e.g., pericyte coverage) as well as serum or plasma biomarkers (e.g., non-coding RNAs). Finally, we highlight the recent advances in combination strategies with the aim of enhancing the response to antiangiogenic therapy in cancer patients and mouse models. This review introduces a comprehensive understanding of the mechanisms and biomarkers associated with the evasion of antiangiogenic therapy in cancer, providing an outlook for developing more effective approaches to promote the therapeutic efficacy of antiangiogenic therapy.
Collapse
Affiliation(s)
- Maohua Huang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yuning Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Chenran Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Lijuan Deng
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Minfeng Chen
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for Biotechnology, St. John's University, NY 11439, USA.
| | - Wencai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
17
|
Orlistat Resensitizes Sorafenib-Resistance in Hepatocellular Carcinoma Cells through Modulating Metabolism. Int J Mol Sci 2022; 23:ijms23126501. [PMID: 35742944 PMCID: PMC9223797 DOI: 10.3390/ijms23126501] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 02/04/2023] Open
Abstract
Sorafenib is one of the options for advanced hepatocellular carcinoma treatment and has been shown to extend median overall survival. However, sorafenib resistance often develops a few months after treatment. Hence, developing various strategies to overcome sorafenib resistance and understand the possible mechanisms is urgently needed. We first established sorafenib-resistant hepatocellular carcinoma (HCC) cells. Then, we found that sorafenib-resistant Huh7 cells (Huh7/SR) exhibit higher glucose uptakes and express elevated fatty acid synthesis and glucose metabolism-related proteins than their parental counterparts (Huh7). The current study investigated whether sorafenib resistance could be reversed by suppressing fatty acid synthesis, using a fatty acid synthase (FASN) inhibitor, orlistat, in HCC cells. FASN inhibition-caused changes in protein expressions and cell cycle distribution were analyzed by Western blot and flow cytometry, and changes in glucose uptakes were also evaluated by 18F-FDG uptake. Orlistat remarkably enhanced the cytotoxicity of sorafenib in both Huh7 and Huh7/SR cells, and flow cytometry showed that combination treatment significantly increased the sub-G1 population in both cell lines. Western blot revealed that the combination treatment effectively increased the ratio of Bax/Bcl-2 and decreased expressions of pERK; additionally, the combination treatment also strongly suppressed fatty acid synthesis-related proteins (e.g., FASN and SCD) in both cell lines. Lastly, the 18F-FDG uptake was repressed by the combination treatment in both cell lines. Our results indicated that orlistat-mediated FASN inhibition could overcome sorafenib resistance and enhance cell killing in HCC by changing cell metabolism.
Collapse
|
18
|
Xiong S, Liu W. Role of metformin in the diagnosis, prevention, and treatment of hepatocellular carcinoma. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:364-373. [PMID: 35545330 PMCID: PMC10930065 DOI: 10.11817/j.issn.1672-7347.2022.210118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Indexed: 06/15/2023]
Abstract
Hepatocellular carcinoma is one of the most common malignant tumors in the world. Although there are many options for the treatment of hepatocellular carcinoma, such as surgical resection, interventional therapy, radiotherapy, chemotherapy, targeted therapy and liver transplantation, the poor therapeutic effect seriously reduces the quality of life for patients and also increases the social and economic burden. Metformin is originally used as the first-line drug for type 2 diabetes, but it has been found to play a certain effect in the prevention and treatment of malignant tumor. The potential roles of metformin against hepatocellular carcinoma, such as regulation of the microenvironment, proliferation signal pathway, metabolism, invasion and metastasis, apoptosis, autophagy, and epigenetics of hepatoma cells. It provides a new choice for the prevention and treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Shize Xiong
- First College of Clinical Medical Science, China Three Gorges University; Institute of Digestive Disease, China Three Gorges University; Department of Gastroenterology, Yichang Central People's Hospital, Yichang Hubei 443000, China.
| | - Wei Liu
- First College of Clinical Medical Science, China Three Gorges University; Institute of Digestive Disease, China Three Gorges University; Department of Gastroenterology, Yichang Central People's Hospital, Yichang Hubei 443000, China.
| |
Collapse
|
19
|
Zeng Z, Lu Q, Liu Y, Zhao J, Zhang Q, Hu L, Shi Z, Tu Y, Xiao Z, Xu Q, Huang D. Effect of the Hypoxia Inducible Factor on Sorafenib Resistance of Hepatocellular Carcinoma. Front Oncol 2021; 11:641522. [PMID: 34307125 PMCID: PMC8292964 DOI: 10.3389/fonc.2021.641522] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/16/2021] [Indexed: 12/11/2022] Open
Abstract
Sorafenib a multi-target tyrosine kinase inhibitor, is the first-line drug for treating advanced hepatocellular carcinoma (HCC). Mechanistically, it suppresses tumor angiogenesis, cell proliferation and promotes apoptosis. Although sorafenib effectively prolongs median survival rates of patients with advanced HCC, its efficacy is limited by drug resistance in some patients. In HCC, this resistance is attributed to multiple complex mechanisms. Previous clinical data has shown that HIFs expression is a predictor of poor prognosis, with further evidence demonstrating that a combination of sorafenib and HIFs-targeted therapy or HIFs inhibitors can overcome HCC sorafenib resistance. Here, we describe the molecular mechanism underlying sorafenib resistance in HCC patients, and highlight the impact of hypoxia microenvironment on sorafenib resistance.
Collapse
Affiliation(s)
- Zhi Zeng
- The Medical College of Qingdao University, Qingdao, China.,Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| | - Qiliang Lu
- The Medical College of Qingdao University, Qingdao, China.,Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| | - Yang Liu
- The Medical College of Qingdao University, Qingdao, China.,Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| | - Junjun Zhao
- Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China.,Graduate Department, Bengbu Medical College, Bengbu, China
| | - Qian Zhang
- The Medical College of Qingdao University, Qingdao, China
| | - Linjun Hu
- The Medical College of Qingdao University, Qingdao, China.,Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| | - Zhan Shi
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yifeng Tu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zunqiang Xiao
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiuran Xu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| | - Dongsheng Huang
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| |
Collapse
|
20
|
Harati R, Vandamme M, Blanchet B, Bardin C, Praz F, Hamoudi RA, Desbois-Mouthon C. Drug-Drug Interaction between Metformin and Sorafenib Alters Antitumor Effect in Hepatocellular Carcinoma Cells. Mol Pharmacol 2021; 100:32-45. [PMID: 33990407 DOI: 10.1124/molpharm.120.000223] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/09/2021] [Indexed: 01/21/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and is one of the leading causes of cancer-related deaths worldwide. The multitarget inhibitor sorafenib is a first-line treatment of patients with advanced unresectable HCC. Recent clinical studies have evidenced that patients treated with sorafenib together with the antidiabetic drug metformin have a survival disadvantage compared with patients receiving sorafenib only. Here, we examined whether a clinically relevant dose of metformin (50 mg/kg per day) could influence the antitumoral effects of sorafenib (15 mg/kg per day) in a subcutaneous xenograft model of human HCC growth using two different sequences of administration, i.e., concomitant versus sequential dosing regimens. We observed that the administration of metformin 6 hours prior to sorafenib was significantly less effective in inhibiting tumor growth (15.4% tumor growth inhibition) than concomitant administration of the two drugs (59.5% tumor growth inhibition). In vitro experiments confirmed that pretreatment of different human HCC cell lines with metformin reduced the effects of sorafenib on cell viability, proliferation, and signaling. Transcriptomic analysis confirmed significant differences between xenografted tumors obtained under the concomitant and the sequential dosing regimens. Taken together, these observations call into question the benefit of parallel use of metformin and sorafenib in patients with advanced HCC and diabetes, as the interaction between the two drugs could ultimately compromise patient survival. SIGNIFICANCE STATEMENT: When drugs are administered sequentially, metformin alters the antitumor effect of sorafenib, the reference treatment for advanced hepatocellular carcinoma, in a preclinical murine xenograft model of liver cancer progression as well as in hepatic cancer cell lines. Defective activation of the AMP-activated protein kinase pathway as well as major transcriptomic changes are associated with the loss of the antitumor effect. These results echo recent clinical work reporting a poorer prognosis for patients with liver cancer who were cotreated with metformin and sorafenib.
Collapse
Affiliation(s)
- Rania Harati
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy (R.H.), and Department of Clinical Sciences, College of Medicine (R.A.H), University of Sharjah, Sharjah, United Arab Emirates; Centre de Recherche Saint-Antoine (R.H., M.V., F.P., C.D.-M.) and Centre de Recherche des Cordeliers (C.D.-M.), Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, Paris, France; Département de Pharmacocinétique et Pharmacochimie, Hôpital Cochin, AP-HP, CARPEM, Paris, France (B.B., C.B.); UMR8038 CNRS, U1268 INSERM, Faculté de Pharmacie, Université de Paris, PRES Sorbonne Paris Cité, Paris, France (B.B); Centre National de la Recherche Scientifique, Paris, France (F.P.); and Division of Surgery and Interventional Science, UCL, London, United Kingdom (R.A.H.)
| | - Marc Vandamme
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy (R.H.), and Department of Clinical Sciences, College of Medicine (R.A.H), University of Sharjah, Sharjah, United Arab Emirates; Centre de Recherche Saint-Antoine (R.H., M.V., F.P., C.D.-M.) and Centre de Recherche des Cordeliers (C.D.-M.), Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, Paris, France; Département de Pharmacocinétique et Pharmacochimie, Hôpital Cochin, AP-HP, CARPEM, Paris, France (B.B., C.B.); UMR8038 CNRS, U1268 INSERM, Faculté de Pharmacie, Université de Paris, PRES Sorbonne Paris Cité, Paris, France (B.B); Centre National de la Recherche Scientifique, Paris, France (F.P.); and Division of Surgery and Interventional Science, UCL, London, United Kingdom (R.A.H.)
| | - Benoit Blanchet
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy (R.H.), and Department of Clinical Sciences, College of Medicine (R.A.H), University of Sharjah, Sharjah, United Arab Emirates; Centre de Recherche Saint-Antoine (R.H., M.V., F.P., C.D.-M.) and Centre de Recherche des Cordeliers (C.D.-M.), Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, Paris, France; Département de Pharmacocinétique et Pharmacochimie, Hôpital Cochin, AP-HP, CARPEM, Paris, France (B.B., C.B.); UMR8038 CNRS, U1268 INSERM, Faculté de Pharmacie, Université de Paris, PRES Sorbonne Paris Cité, Paris, France (B.B); Centre National de la Recherche Scientifique, Paris, France (F.P.); and Division of Surgery and Interventional Science, UCL, London, United Kingdom (R.A.H.)
| | - Christophe Bardin
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy (R.H.), and Department of Clinical Sciences, College of Medicine (R.A.H), University of Sharjah, Sharjah, United Arab Emirates; Centre de Recherche Saint-Antoine (R.H., M.V., F.P., C.D.-M.) and Centre de Recherche des Cordeliers (C.D.-M.), Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, Paris, France; Département de Pharmacocinétique et Pharmacochimie, Hôpital Cochin, AP-HP, CARPEM, Paris, France (B.B., C.B.); UMR8038 CNRS, U1268 INSERM, Faculté de Pharmacie, Université de Paris, PRES Sorbonne Paris Cité, Paris, France (B.B); Centre National de la Recherche Scientifique, Paris, France (F.P.); and Division of Surgery and Interventional Science, UCL, London, United Kingdom (R.A.H.)
| | - Françoise Praz
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy (R.H.), and Department of Clinical Sciences, College of Medicine (R.A.H), University of Sharjah, Sharjah, United Arab Emirates; Centre de Recherche Saint-Antoine (R.H., M.V., F.P., C.D.-M.) and Centre de Recherche des Cordeliers (C.D.-M.), Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, Paris, France; Département de Pharmacocinétique et Pharmacochimie, Hôpital Cochin, AP-HP, CARPEM, Paris, France (B.B., C.B.); UMR8038 CNRS, U1268 INSERM, Faculté de Pharmacie, Université de Paris, PRES Sorbonne Paris Cité, Paris, France (B.B); Centre National de la Recherche Scientifique, Paris, France (F.P.); and Division of Surgery and Interventional Science, UCL, London, United Kingdom (R.A.H.)
| | - Rifat Akram Hamoudi
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy (R.H.), and Department of Clinical Sciences, College of Medicine (R.A.H), University of Sharjah, Sharjah, United Arab Emirates; Centre de Recherche Saint-Antoine (R.H., M.V., F.P., C.D.-M.) and Centre de Recherche des Cordeliers (C.D.-M.), Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, Paris, France; Département de Pharmacocinétique et Pharmacochimie, Hôpital Cochin, AP-HP, CARPEM, Paris, France (B.B., C.B.); UMR8038 CNRS, U1268 INSERM, Faculté de Pharmacie, Université de Paris, PRES Sorbonne Paris Cité, Paris, France (B.B); Centre National de la Recherche Scientifique, Paris, France (F.P.); and Division of Surgery and Interventional Science, UCL, London, United Kingdom (R.A.H.)
| | - Christèle Desbois-Mouthon
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy (R.H.), and Department of Clinical Sciences, College of Medicine (R.A.H), University of Sharjah, Sharjah, United Arab Emirates; Centre de Recherche Saint-Antoine (R.H., M.V., F.P., C.D.-M.) and Centre de Recherche des Cordeliers (C.D.-M.), Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, Paris, France; Département de Pharmacocinétique et Pharmacochimie, Hôpital Cochin, AP-HP, CARPEM, Paris, France (B.B., C.B.); UMR8038 CNRS, U1268 INSERM, Faculté de Pharmacie, Université de Paris, PRES Sorbonne Paris Cité, Paris, France (B.B); Centre National de la Recherche Scientifique, Paris, France (F.P.); and Division of Surgery and Interventional Science, UCL, London, United Kingdom (R.A.H.)
| |
Collapse
|
21
|
Tang Z, Tang N, Jiang S, Bai Y, Guan C, Zhang W, Fan S, Huang Y, Lin H, Ying Y. The Chemosensitizing Role of Metformin in Anti-Cancer Therapy. Anticancer Agents Med Chem 2021; 21:949-962. [PMID: 32951587 DOI: 10.2174/1871520620666200918102642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/23/2020] [Accepted: 08/08/2020] [Indexed: 11/22/2022]
Abstract
Chemoresistance, which leads to the failure of chemotherapy and further tumor recurrence, presents the largest hurdle for the success of anti-cancer therapy. In recent years, metformin, a widely used first-line antidiabetic drug, has attracted increasing attention for its anti-cancer effects. A growing body of evidence indicates that metformin can sensitize tumor responses to different chemotherapeutic drugs, such as hormone modulating drugs, anti-metabolite drugs, antibiotics, and DNA-damaging drugs via selective targeting of Cancer Stem Cells (CSCs), improving the hypoxic microenvironment, and by suppressing tumor metastasis and inflammation. In addition, metformin may regulate metabolic programming, induce apoptosis, reverse Epithelial to Mesenchymal Transition (EMT), and Multidrug Resistance (MDR). In this review, we summarize the chemosensitization effects of metformin and focus primarily on its molecular mechanisms in enhancing the sensitivity of multiple chemotherapeutic drugs, through targeting of mTOR, ERK/P70S6K, NF-κB/HIF-1 α, and Mitogen- Activated Protein Kinase (MAPK) signaling pathways, as well as by down-regulating the expression of CSC genes and Pyruvate Kinase isoenzyme M2 (PKM2). Through a comprehensive understanding of the molecular mechanisms of chemosensitization provided in this review, the rationale for the use of metformin in clinical combination medications can be more systematically and thoroughly explored for wider adoption against numerous cancer types.>.
Collapse
Affiliation(s)
- Zhimin Tang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Nan Tang
- Nanchang Joint Program, Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Shanshan Jiang
- Institute of Hematological Research, Shanxi Provincial People's Hospital, Xian 710000, China
| | - Yangjinming Bai
- Nanchang Joint Program, Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Chenxi Guan
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Wansi Zhang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Shipan Fan
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510005, China
| | - Yonghong Huang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Hui Lin
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| |
Collapse
|
22
|
Chen YC, Li H, Wang J. Mechanisms of metformin inhibiting cancer invasion and migration. Am J Transl Res 2020; 12:4885-4901. [PMID: 33042396 PMCID: PMC7540116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/01/2020] [Indexed: 06/11/2023]
Abstract
Cancer currently ranks among the leading causes of death globally. Cancer invasion and metastasis transform locally grown cancers to a systemic and life-threatening disease, which accounts for the most significant challenge in cancer treatment. Recent studies showed that Metformin, the most commonly used first-line oral drug for the treatment of type 2 diabetes (T2DM), could prevent and treat various cancers. Moreover, multiple evidence suggested that metformin inhibited cancer invasion and metastasis, which could improve the prognosis of cancer patients administrated with metformin. To better understand the anti-cancer role of metformin, the present review summarized the potential mechanisms of inhibiting cancer invasion and metastasis by metformin, including AMPK signaling pathway, EMT signaling pathway, epigenetic modification and so on. However, multiple problems remain unresolved and more clinical trials are needed to prove the inhibition of cancer invasion and metastasis by metformin.
Collapse
Affiliation(s)
- Yong Chang Chen
- Gynecologic Oncology Clinical Research Center, Hunan Cancer Hospital, Central South UniversityChangsha 410013, Hunan, China
- University of South ChinaHengyang 421001, Hunan, China
| | - He Li
- Gynecologic Oncology Clinical Research Center, Hunan Cancer Hospital, Central South UniversityChangsha 410013, Hunan, China
| | - Jing Wang
- Gynecologic Oncology Clinical Research Center, Hunan Cancer Hospital, Central South UniversityChangsha 410013, Hunan, China
| |
Collapse
|
23
|
Wang B, Dong J, Xiao H, Li Y, Jin Y, Cui M, Zhang SQ, Fan SJ. Metformin fights against radiation-induced early developmental toxicity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 732:139274. [PMID: 32438158 DOI: 10.1016/j.scitotenv.2020.139274] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/20/2020] [Accepted: 05/06/2020] [Indexed: 06/11/2023]
Abstract
Nuclear pollution intertwined accidental irradiation not only triggers acute and chronic radiation syndromes, but also endangers embryonic development in sight of uncontrollable gene mutation. Metformin (MET), a classic hypoglycemic drug, has been identified to possess multiple properties. In this study, we explored the radioprotective effects of MET on the developmental abnormalities and deformities induced by irradiation among three "star drugs". Specifically, zebrafish (Danio rerio) embryos exposed to 5.2 Gy gamma irradiation at 4 h post fertilization (hpf) showed overt developmental toxicity, including hatching delay, hatching rate decrease, developmental indexes reduction, morphological abnormalities occurrence and motor ability decline. However, MET treatment erased the radiation-induced phenotypes. In addition, MET degraded inflammatory reaction, hinders apoptosis response, and reprograms the development-related genes expression, such as sox2, sox3, sox19a and p53, in zebrafish embryos following radiation challenge. Together, our findings provide novel insights into metformin, and underpin that metformin might be employed as a promising radioprotector for radiation-induced early developmental toxicity in pre-clinical settings.
Collapse
Affiliation(s)
- Bin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiali Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Huiwen Xiao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yuxiao Jin
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.
| | - Shu-Qin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.
| | - Sai-Jun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.
| |
Collapse
|
24
|
Sang J, Tang R, Yang M, Sun Q. Metformin Inhibited Proliferation and Metastasis of Colorectal Cancer and presented a Synergistic Effect on 5-FU. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9312149. [PMID: 32851092 PMCID: PMC7439187 DOI: 10.1155/2020/9312149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022]
Abstract
The purpose of this study was to investigate the effect of metformin or the combination of metformin and 5-FU on the growth and metastasis of colorectal cancer (CRC). For the in vitro experiments, HCT 116 and SW1463 cell lines were treated with metformin or the combination of metformin and 5-FU. Cell proliferation and invasion were analyzed by CCK-8, colony formation, and transwell assay, respectively. For the in vivo experiments, the CRC xenograft nude mice model was used to observe the effects of metformin or combined with 5-FU on tumor growth and metastasis. Metformin significantly inhibited the proliferation and invasion of HCT116 and SW1463 cells in vitro, which showed synergetic effects to 5-FU. In CRC xenograft nude mice, metformin alone and metformin combined with 5-FU treatment significantly inhibited tumor cell proliferation and tumor metastasis. In summary, metformin played an inhibitory role in the proliferation and metastasis of CRC and had a synergistic effect with 5-FU. Metformin may be a potentially effective anti-metastatic drug or an anticancer adjuvant agent for treating CRC.
Collapse
Affiliation(s)
- Jing Sang
- Department of Pathology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250014, China
- Department of Pathology, Tai'an Central Hospital, Tai'an 271000, China
| | - Ruixue Tang
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| | - Min Yang
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| | - Qing Sun
- Department of Pathology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250014, China
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| |
Collapse
|
25
|
Sorafenib and Mesenchymal Stem Cell Therapy: A Promising Approach for Treatment of HCC. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9602728. [PMID: 32617114 PMCID: PMC7312705 DOI: 10.1155/2020/9602728] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/06/2020] [Accepted: 05/21/2020] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most commonly diagnosed cancer and the second most common cause of cancer-related death worldwide. Sorafenib (Sora) is used as a targeted therapy for HCC treatment. Mesenchymal stem cells (MSCs) are applied as a new approach to fight malignancies. Drug resistance and side effects are the major concerns with Sora administration. The effect of using the combination of sorafenib and MSCs on tumor regression in xenograft HCC models was evaluated in this study. Methods and Materials. Human hepatocellular carcinoma cell lines (HepG2) were subcutaneously implanted into the flank of 18 nude mice. The animals were randomly divided into six groups (n = 3); each received Sora (oral), MSCs (IV injection), MSCs (local injection), Sora + MSCs (IV injection), Sora + MSCs (local injection), or no treatment (the control group). Six weeks after tumor implantation, the mice were scarified and tumoral tissues were resected in their entirety. Histopathological and immunohistochemical evaluations were used to measure tumor proliferation and angiogenesis. Apoptotic cells were quantified using the TUNEL assay. Results. No significant difference was found in the tumor grade among the treatment groups. Differentiation features of the tumoral cells were histopathologically insignificant in all the groups. Tumor necrosis was highest in the hpMSC (local) + Sora group. Tumor cell proliferation was reduced in hpMSC (local) + Sora-treated and hpMSC (IV) + Sora-treated mice compared with the other groups. Apoptotic-positive cells occupied a greater proportion in the Sora, hpMSC (IV) + Sora, and hpMSC (local) + Sora groups. Conclusion. A combination of chemotherapy and MSC can yield to more favorable results in the treatment of HCC.
Collapse
|
26
|
Li M, Li J, Guo X, Pan H, Zhou Q. Absence of HTATIP2 Expression in A549 Lung Adenocarcinoma Cells Promotes Tumor Plasticity in Response to Hypoxic Stress. Cancers (Basel) 2020; 12:cancers12061538. [PMID: 32545251 PMCID: PMC7352940 DOI: 10.3390/cancers12061538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022] Open
Abstract
HIV-1 Tat Interactive Protein 2 (HTATIP2) is a tumor suppressor, of which reduced or absent expression is associated with increased susceptibility to tumorigenesis and enhanced tumor invasion and metastasis. However, whether the absent expression of HTATIP2 is a tumor-promoting factor that acts through improving tumor adaptation to hypoxia is unclear. Here, we established a stable HTATIP2-knockdown A549 human lung adenocarcinoma cell line (A549shHTATIP2) using lentiviral-delivered HTATIP2-targeting short hairpin RNA (shRNA), employed a double subcutaneous xenograft model and incorporated photoacoustic imaging and metabolomics approaches to elucidate the impact of the absent HTATIP2 expression on tumor response to hypoxic stress. Results from the in vivo study showed that A549shHTATIP2 tumors exhibited accelerated growth but decreased intratumoral oxygenation and angiogenesis and reduced sensitivity to sorafenib treatment as compared with their parental counterparts. Moreover, results of the immunoblot and real-time PCR analyses revealed that the HIF2α protein and mRNA levels in vehicle-treated A549shHTATIP2 tumors were significantly increased (p < 0.01 compared with the parental control tumors). Despite the strong HIF2α-c-Myc protein interaction indicated by our co-immunoprecipitation data, the increase in the c-Myc protein and mRNA levels was not significant in the A549shHTATIP2 tumors. Nonetheless, MCL-1 and β-catenin protein levels in A549shHTATIP2 tumors were significantly increased (p < 0.05 compared with the parental control tumors), suggesting an enhanced β-catenin/c-Myc/MCL-1 pathway in the absence of HTATIP2 expression. The finding of significantly decreased E-cadherin (p < 0.01 compared with vehicle-treated A549shHTATIP2 tumors) and increased vimentin (p < 0.05 compared with sorafenib-treated A549 tumors) protein levels in A549shHTATIP2 tumors implicates that the absence of HTATIP2 expression increases the susceptibility of A549 tumors to sorafenib-activated epithelial-mesenchymal transition (EMT) process. Comparison of the metabolomic profiles between A549 and A549shHTATIP2 tumors demonstrated that the absence of HTATIP2 expression resulted in increased tumor metabolic plasticity that enabled tumor cells to exploit alternative metabolic pathways for survival and proliferation rather than relying on glutamine and fatty acids as a carbon source to replenish TCA cycle intermediates. Our data suggest a mechanism by which the absent HTATIP2 expression modulates tumor adaptation to hypoxia and promotes an aggressive tumor phenotype by enhancing the HIF2α-regulated β-catenin/c-Myc/MCL-1 signaling, increasing the susceptibility of tumors to sorafenib treatment-activated EMT process, and improving tumor metabolic plasticity.
Collapse
Affiliation(s)
- Minghua Li
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (M.L.); (X.G.)
| | - Jing Li
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Xiaofang Guo
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (M.L.); (X.G.)
| | - Hua Pan
- Division of Cardiovascular Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Qingyu Zhou
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (M.L.); (X.G.)
- Correspondence: ; Tel.: +1-813-974-7081
| |
Collapse
|
27
|
Malale K, Fu J, Qiu L, Zhan K, Gan X, Mei Z. Hypoxia-Induced Aquaporin-3 Changes Hepatocellular Carcinoma Cell Sensitivity to Sorafenib by Activating the PI3K/Akt Signaling Pathway. Cancer Manag Res 2020; 12:4321-4333. [PMID: 32606928 PMCID: PMC7294049 DOI: 10.2147/cmar.s243918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/12/2020] [Indexed: 12/29/2022] Open
Abstract
Purpose Hypoxia-induced changes are primarily activated in patients with hepatocellular carcinoma (HCC) and long-term sorafenib exposure, thereby reducing the sensitivity to the drug. Aquaporin-3 (AQP3), a member of the aquaporin family, is a hypoxia-induced substance that affects the chemosensitivity of non-hepatocellular tumors. However, its expression and role in the sensitivity of hypoxic HCC cells to sorafenib-induced apoptosis remain unclear. The purpose of this study was to detect changes in AQP3 expression in hypoxic HCC cells and to determine whether these changes alter the sensitivity of these cells to sorafenib. Materials and Methods Huh7 and HepG2 hypoxic cell models were established and AQP3 expression was detected using quantitative real-time polymerase chain reaction (qPCR) and Western blotting. Furthermore, the role of AQP3 in cell sensitivity to sorafenib was evaluated via flow cytometry, Western blotting, and a CCK-8 assay. Results The results of qPCR and Western blotting showed that AQP3 was overexpressed in the Huh7 and HepG2 hypoxic cell models. Furthermore, AQP3 protein levels were positively correlated with hypoxia-inducible factor-1α (HIF-1α) levels. Compared with cells transfected with lentivirus-GFP (Lv-GFP), hypoxic cells transfected with lentivirus-AQP3 (Lv-AQP3) were less sensitive to sorafenib-induced apoptosis. However, the sensitivity to the drug increased in cells transfected with lentivirus-AQP3RNAi (Lv-AQP3RNAi). Akt and Erk phosphorylation was enhanced in Lv-AQP3-transfected cells. Compared with UO126 (a Mek1/2 inhibitor), LY294002 (a PI3K inhibitor) attenuated the AQP3-induced insensitivity to sorafenib observed in hypoxic cells transfected with Lv-AQP3. Combined with LY294002-treated cells, hypoxic cells transfected with Lv-AQP3RNAi were more sensitive to sorafenib. Conclusion The study results show that AQP3 is a potential therapeutic target for improving the sensitivity of hypoxic HCC cells to sorafenib.
Collapse
Affiliation(s)
- Kija Malale
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jili Fu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Liewang Qiu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Ke Zhan
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xiuni Gan
- Department of Nursing, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhechuan Mei
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
28
|
Fan G, Wei X, Xu X. Is the era of sorafenib over? A review of the literature. Ther Adv Med Oncol 2020; 12:1758835920927602. [PMID: 32518599 PMCID: PMC7252361 DOI: 10.1177/1758835920927602] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 04/27/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most severe diseases worldwide. For the different stages of HCC, there are different clinical treatment strategies, such as surgical therapy for the early stage, and transarterial chemoembolization (TACE) and selective internal radiation therapy (SIRT) for intermediate-stage disease. Systemic treatment, which uses mainly targeted drugs, is the standard therapy against advanced HCC. Sorafenib is an important first-line therapy for advanced HCC. As a classically effective drug, sorafenib can increase overall survival markedly. However, it still has room for improvement because of the heterogeneity of HCC and acquired resistance. Scientists have reported the acquired sorafenib resistance is associated with the anomalous expression of certain genes, most of which are also related with HCC onset and development. Combining sorafenib with inhibitors targeting these genes may be an effective treatment. Combined treatment may not only overcome drug resistance, but also inhibit the expression of carcinoma-related genes. This review focuses on the current status of sorafenib in advanced HCC, summarizes the inhibitors that can combine with sorafenib in the treatment against HCC, and provides the rationale for clinical trials of sorafenib in combination with other inhibitors in HCC. The era of sorafenib in the treatment of HCC is far from over, as long as we find better methods of medication.
Collapse
Affiliation(s)
- Guanghan Fan
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-organ Transplantation; Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-organ Transplantation; Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-organ Transplantation; Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS; Key Laboratory of Organ Transplantation, Zhejiang Province, 79 QingChun Road, Hangzhou, 310003, China
| |
Collapse
|
29
|
Yu XN, Deng Y, Zhang GC, Liu J, Liu TT, Dong L, Zhu CF, Shen XZ, Li YH, Zhu JM. Sorafenib-Conjugated Zinc Phthalocyanine Based Nanocapsule for Trimodal Therapy in an Orthotopic Hepatocellular Carcinoma Xenograft Mouse Model. ACS APPLIED MATERIALS & INTERFACES 2020; 12:17193-17206. [PMID: 32207914 DOI: 10.1021/acsami.0c00375] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Sorafenib, a multitargeted kinase inhibitor, has been reported to elicit a limited therapeutic effect in hepatocellular carcinoma (HCC). Currently, phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), is emerging as a powerful modality for cancer therapy. However, few studies have been reported the effectiveness of the combination of sorafenib with PDT and PTT in HCC. Herein, we designed and synthesized bovine serum albumin (BSA)-coated zinc phthalocyanine (ZnPc) and sorafenib (SFB) nanoparticle (ZnPc/SFB@BSA). The obtained ZnPc/SFB@BSA was able to trigger PDT, PTT, and chemotherapy. After irradiation by a 730 nm light, ZnPc/SFB@BSA significantly suppressed HCC cell proliferation and metastasis while promoted cell apoptosis in vitro. Furthermore, intravenous injection of ZnPc/SFB@BSA led to dramatically reduced tumor growth in an orthotopic xenograft HCC model. More importantly, ZnPc/SFB@BSA presented low toxicity and adequate blood compatibility. Therefore, a combination of ZnPc with sorafenib via BSA-assembled nanoparticle can markedly suppress HCC growth, representing a promising strategy for HCC patients.
Collapse
Affiliation(s)
- Xiang-Nan Yu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Shanghai Institute of Liver Disease, Shanghai 200032, China
| | - Yong Deng
- Institute of Bismuth Science & College of Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Guang-Cong Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Shanghai Institute of Liver Disease, Shanghai 200032, China
| | - Jie Liu
- Institute of Bismuth Science & College of Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Tao-Tao Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Shanghai Institute of Liver Disease, Shanghai 200032, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Shanghai Institute of Liver Disease, Shanghai 200032, China
| | - Chang-Feng Zhu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Shanghai Institute of Liver Disease, Shanghai 200032, China
| | - Xi-Zhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Shanghai Institute of Liver Disease, Shanghai 200032, China
- Key Laboratory of Medical Molecular Virology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yu-Hao Li
- Institute of Bismuth Science & College of Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Ji-Min Zhu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Shanghai Institute of Liver Disease, Shanghai 200032, China
| |
Collapse
|
30
|
Cao MQ, You AB, Cui W, Zhang S, Guo ZG, Chen L, Zhu XD, Zhang W, Zhu XL, Guo H, Deng DJ, Sun HC, Zhang T. Cross talk between oxidative stress and hypoxia via thioredoxin and HIF-2α drives metastasis of hepatocellular carcinoma. FASEB J 2020; 34:5892-5905. [PMID: 32157720 DOI: 10.1096/fj.202000082r] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/25/2020] [Accepted: 02/25/2020] [Indexed: 12/30/2022]
Abstract
Oxidative stress and hypoxia are two opposite microenvironments involved in HCC metastasis. Thioredoxin (TXN) and hypoxia-inducible factor 2α (HIF-2α) are typical proteins involved in these two different microenvironments, respectively. How these two factors interact to influence the fate on tumor cells remains unknown. Hypoxia facilitated HCC cells withstood oxidative stress and eventually promoted HCC cells metastasis, in which TXN and HIF-2α were mostly involved. Upregulation of TXN/HIF-2α correlated with poor HCC prognosis and promoted HCC metastasis both in vitro and in vivo. Epithelial-mesenchymal transition (EMT) process was involved in TXN/HIF-2α-enhanced invasiveness of HCC cells. Additionally, the stability and activity of HIF-2α were precisely regulated by TXN via SUMOylation and acetylation, which contributed to HCC metastasis. Our data revealed that the redox protein TXN and HIF-2α are both associated with HCC metastasis, and the fine regulation of TXN on HIF-2α contributes essentially during the process of metastasis. Our study provides new insight into the interaction mechanism between hypoxia and oxidative stress and implies potential therapeutic benefits by targeting both TXN and HIF-2α in the treatment of HCC metastasis.
Collapse
Affiliation(s)
- Man-Qing Cao
- Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - A-Bin You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wei Cui
- Key Laboratory of Artificial Cell, Institute for Hepatobiliary Diseases, Tianjin Third Central Hospital, Tianjin, China
| | - Su Zhang
- Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhi-Gui Guo
- Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Lu Chen
- Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiao-Dong Zhu
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiao-Lin Zhu
- Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hua Guo
- Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Da-Jun Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hui-Chuan Sun
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ti Zhang
- Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
31
|
Ma W, Chen X, Wu X, Li J, Mei C, Jing W, Teng L, Tu H, Jiang X, Wang G, Chen Y, Wang K, Wang H, Wei Y, Liu Z, Yuan Y. Long noncoding RNA SPRY4-IT1 promotes proliferation and metastasis of hepatocellular carcinoma via mediating TNF signaling pathway. J Cell Physiol 2020; 235:7849-7862. [PMID: 31943198 DOI: 10.1002/jcp.29438] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 12/23/2019] [Indexed: 01/17/2023]
Abstract
Our previous studies have indicated that long noncoding RNA (lncRNA) SPRY4 intronic transcript 1 (SPRY4-IT1) was highly expressed in hepatocellular carcinoma (HCC). However, it still remained unclear how SPRY4-IT1 worked in tumorgenesis in HCC. In this study, we tested the overexpression of SPRY4-IT1 in HCC tissues and cells through a quantitative real-time polymerase chain reaction. Statistical analyses showed that the upregulation had an association with the tumor node metastasis stage, thrombin time, and alkaline phosphatase. Furthermore, SPRY4-IT1 could be involved in cell proliferation, metastasis, and the epithelial-to-mesenchymal transition (EMT) process in HCC in vitro and in vivo. RNA-sequencing and transcriptome analysis were carried out to explore the mechanism of SPRY4-IT1 in HCC. With SPRY4-IT1 being knocked down or overexpressed, the level of proteins in the tumor necrosis factor (TNF) signaling pathway changed. We detected the RNA binding protein heterogeneous nuclear ribonucleoprotein L (HNRNPL) as a SPRY4-IT1 interacting protein through RNA pull-down assay and liquid chromatography-mass spectrometry, then verified through RNA immunoprecipitation. Downregulation of HNRNPL induced the change of proteins observed on SPRY4-IT1 downregulation revealing the SPRY4-IT1: HNRNPL complex in the TNF signaling pathway and EMT process in HCC. In general, our experimental data and analysis demonstrated the role of SPRY4-IT1 in promoting progress and metastasis of HCC by the TNF signaling pathway.
Collapse
Affiliation(s)
- Weijie Ma
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xi Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaoling Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jinghua Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chengjie Mei
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Jing
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Teng
- Department of Pathology, Wuhan Women and Children Medical Care Center, Wuhan, China
| | - Honglei Tu
- Department of Clinical Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiang Jiang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ganggang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yiran Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kunlei Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Haitao Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhisu Liu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
32
|
Méndez-Blanco C, Fondevila F, Fernández-Palanca P, García-Palomo A, van Pelt J, Verslype C, González-Gallego J, Mauriz JL. Stabilization of Hypoxia-Inducible Factors and BNIP3 Promoter Methylation Contribute to Acquired Sorafenib Resistance in Human Hepatocarcinoma Cells. Cancers (Basel) 2019; 11:E1984. [PMID: 31835431 PMCID: PMC6966438 DOI: 10.3390/cancers11121984] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/27/2019] [Accepted: 12/06/2019] [Indexed: 01/19/2023] Open
Abstract
Despite sorafenib effectiveness against advanced hepatocarcinoma (HCC), long-term exposure to antiangiogenic drugs leads to hypoxic microenvironment, a key contributor to chemoresistance acquisition. We aimed to study the role of hypoxia in the development of sorafenib resistance in a human HCC in vitro model employing the HCC line HepG2 and two variants with acquired sorafenib resistance, HepG2S1 and HepG2S3, and CoCl2 as hypoximimetic. Resistant cells exhibited a faster proliferative rate and hypoxia adaptive mechanisms, linked to the increased protein levels and nuclear translocation of hypoxia-inducible factors (HIFs). HIF-1α and HIF-2α overexpression was detected even under normoxia through a deregulation of its degradation mechanisms. Proapoptotic markers expression and subG1 population decreased significantly in HepG2S1 and HepG2S3, suggesting evasion of sorafenib-mediated cell death. HIF-1α and HIF-2α knockdown diminished resistant cells viability, relating HIFs overexpression with its prosurvival ability. Additionally, epigenetic silencing of Bcl-2 interacting protein 3 (BNIP3) was observed in sorafenib resistant cells under hypoxia. Demethylation of BNIP3 promoter, but not histone acetylation, restored BNIP3 expression, driving resistant cells' death. Altogether, our results highlight the involvement of HIFs overexpression and BNIP3 methylation-dependent knockdown in the development of sorafenib resistance in HCC. Targeting both prosurvival mechanisms could overcome chemoresistance and improve future therapeutic approaches.
Collapse
Affiliation(s)
- Carolina Méndez-Blanco
- Institute of Biomedicine (IBIOMED), University of León, Campus of Vegazana s/n, 24071 León, Spain; (C.M.-B.); (F.F.); (P.F.-P.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
| | - Flavia Fondevila
- Institute of Biomedicine (IBIOMED), University of León, Campus of Vegazana s/n, 24071 León, Spain; (C.M.-B.); (F.F.); (P.F.-P.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
| | - Paula Fernández-Palanca
- Institute of Biomedicine (IBIOMED), University of León, Campus of Vegazana s/n, 24071 León, Spain; (C.M.-B.); (F.F.); (P.F.-P.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
| | - Andrés García-Palomo
- Service of Oncology, Complejo Asistencial Universitario de León, Calle Altos de Nava, s/n, 24001 León, Spain;
| | - Jos van Pelt
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven and University Hospitals Leuven and Leuven Cancer Institute (LKI), 3000 Leuven, Belgium; (J.v.P.)
| | - Chris Verslype
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven and University Hospitals Leuven and Leuven Cancer Institute (LKI), 3000 Leuven, Belgium; (J.v.P.)
| | - Javier González-Gallego
- Institute of Biomedicine (IBIOMED), University of León, Campus of Vegazana s/n, 24071 León, Spain; (C.M.-B.); (F.F.); (P.F.-P.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
| | - José L. Mauriz
- Institute of Biomedicine (IBIOMED), University of León, Campus of Vegazana s/n, 24071 León, Spain; (C.M.-B.); (F.F.); (P.F.-P.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
| |
Collapse
|
33
|
Kok VC, Chen YC, Chen YY, Su YC, Ku MC, Kuo JT, Yoshida GJ. Sorafenib with Transarterial Chemoembolization Achieves Improved Survival vs. Sorafenib Alone in Advanced Hepatocellular Carcinoma: A Nationwide Population-Based Cohort Study. Cancers (Basel) 2019; 11:985. [PMID: 31311148 PMCID: PMC6679028 DOI: 10.3390/cancers11070985] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/06/2019] [Accepted: 07/12/2019] [Indexed: 02/07/2023] Open
Abstract
We hypothesized that sorafenib plus transarterial chemoembolization (TACE) would confer survival benefits over sorafenib alone for advanced hepatocellular carcinoma (aHCC). We investigated this while using the population-based All-Cancer Dataset to assemble a cohort (n = 3674; median age, 60; 83% men) of patients receiving sorafenib for aHCC (Child-Pugh A) with macro-vascular invasion or nodal/distant metastases. The patients were classified into the sorafenib-TACE group (n = 426) or the propensity score-matched sorafenib-alone group (n = 1686). All of the participants were followed up until death or the end of the study. Time-dependent Cox model and the Mantel-Byar test were used for survival analysis. During the median follow-ups of 221 and 133 days for the sorafenib-TACE and sorafenib-alone groups, 164 (39%) and 916 (54%) deaths occurred, respectively; the corresponding median overall survivals (OS) were 381 and 204 days, respectively (hazard ratio, HR: 0.74; 95% confidence interval, CI, 0.63-0.88; p = 0.021). The one-year and six-month OS were 53.5% and 80.3% in the sorafenib-TACE group and 32.4% and 54.4% in the sorafenib-alone group, respectively. The major complications were comparable between the two groups. The addition of TACE to sorafenib improves survival, with a 26% reduction in mortality. These findings provide strong real-world evidence that supports this combination strategy for eligible Child-Pugh A aHCC patients.
Collapse
Affiliation(s)
- Victor C Kok
- Division of Medical Oncology, Department of Internal Medicine, Kuang Tien General Hospital, Taichung 43303, Taiwan.
- Disease Informatics Research Group, Department of Bioinformatics and Medical Engineering, Asia University Taiwan, Taichung 41354, Taiwan.
- Student, Cancer Biology and Therapeutics: High-Impact Cancer Research Postgraduate Certificate Program, Harvard Medical School, Boston, MA 02115, USA.
| | - Yu-Ching Chen
- Disease Informatics Research Group, Department of Bioinformatics and Medical Engineering, Asia University Taiwan, Taichung 41354, Taiwan
| | - Yang-Yuan Chen
- Department of Gastroenterology, Changhua Christian Medical Foundation Changhua Christian Hospital, Changhua 50006, Taiwan
| | - Yu-Chieh Su
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Ming-Chang Ku
- Interventional Radiology Unit, Department of Radiology, Kuang Tien General Hospital, Taichung 43303, Taiwan
| | - Jung-Tsung Kuo
- Artificial Intelligence Center for Medical Diagnosis, China Medical University Hospital, Taichung 40447, Taiwan
| | - Go J Yoshida
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Faculty of Medical Science, Graduate School Juntendo University, Tokyo 113-8421, Japan
| |
Collapse
|
34
|
Wu Z, Ai X, Hu H, Wang S, Wang Y, Kang F, Ouyang C, Zhu J. Hematopoietic-substrate-1 associated protein X-1 (HAX-1) regulates liver cancer cells growth, metastasis, and angiogenesis through Akt. Cancer Biol Ther 2019; 20:1223-1233. [PMID: 31132019 DOI: 10.1080/15384047.2019.1617562] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The aim of this study was to investigate the effects and mechanisms of hematopoietic-substrate-1-associated protein X-1 (HAX-1) on liver cancer cells. Information on HAX-1 from liver cancer patients was analyzed by the Cancer Genome Atlas (TCGA) program. Cell migration and invasion abilities were respectively tested by scratch assay and transwell assay. Tube formation assay was applied to detect angiogenesis protein and mRNA was determined using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. We found that the median month survival of HAX-1 overexpressing liver cancer patients was shorter than that of HAX-1 normal liver cancer patients. HAX-1 was overexpressed in liver cancer tissues and cells, and HAX-1 overexpression promoted the liver cancer cells growth, migration, and invasion, whereas silencing HAX-1 produced the opposite results. Inhibition of Akt by LY294002 reversed the migration and invasion abilities of liver cancer cells, and inhibited the ability of cells growth and angiogenesis. Silencing PIK3CA enhanced the inhibitory effects of HAX-1 silencing on the viability, migration, and invasion of liver cancer cells. HAX-1 affected liver cancer cells metastasis and angiogenesis by affecting Akt phosphorylation and FOXO3A expression.
Collapse
Affiliation(s)
- Zhenyu Wu
- Department of Hepatobiliary Surgery, Aerospace Center Hospital , Beijing , China
| | - Xiangnan Ai
- Department of Hepatobiliary Surgery, Aerospace Center Hospital , Beijing , China
| | - Hao Hu
- Department of Hepatobiliary Surgery, Aerospace Center Hospital , Beijing , China
| | - Siqi Wang
- Department of Hepatobiliary Surgery, Peking University People's Hospital , Beijing , China
| | - Yang Wang
- Department of Hepatobiliary Surgery, Peking University People's Hospital , Beijing , China
| | - Feng Kang
- Beijing Vitalstar Biotechnology Co., Ltd ., Beijing , China
| | - Caiguo Ouyang
- Department of Hepatobiliary Surgery, Aerospace Center Hospital , Beijing , China
| | - Jiye Zhu
- Department of Hepatobiliary Surgery, Peking University People's Hospital , Beijing , China
| |
Collapse
|
35
|
Jiang X, Tan HY, Teng S, Chan YT, Wang D, Wang N. The Role of AMP-Activated Protein Kinase as a Potential Target of Treatment of Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:647. [PMID: 31083406 PMCID: PMC6562911 DOI: 10.3390/cancers11050647] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is the fifth most frequent cancer worldwide with a very high recurrence rate and very dismal prognosis. Diagnosis and treatment in HCC remain difficult, and the identification of new therapeutic targets is necessary for a better outcome of HCC treatment. AMP-Activated Protein Kinase (AMPK) is an essential intracellular energy sensor that plays multiple roles in cellular physiology and the pathological development of chronic diseases. Recent studies have highlighted the important regulation of AMPK in HCC. This review aims to comprehensively and critically summarize the role of AMPK in HCC. Methods: Original studies were retrieved from NCBI database with keywords including AMPK and HCC, which were analyzed with extensive reading. Results: Dysregulation of the kinase activity and expression of AMPK was observed in HCC, which was correlated with survival of the patients. Loss of AMPK in HCC cells may proceed cell cycle progression, proliferation, survival, migration, and invasion through different oncogenic molecules and pathways. Conclusions: We identified several AMPK activators which may possess potential anti-HCC function, and discussed the clinical perspective on the use of AMPK activators for HCC therapy.
Collapse
Affiliation(s)
- Xue Jiang
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Hor-Yue Tan
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| | - Shanshan Teng
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Yau-Tuen Chan
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
36
|
Bort A, Sánchez BG, Mateos-Gómez PA, Vara-Ciruelos D, Rodríguez-Henche N, Díaz-Laviada I. Targeting AMP-activated kinase impacts hepatocellular cancer stem cells induced by long-term treatment with sorafenib. Mol Oncol 2019; 13:1311-1331. [PMID: 30959553 PMCID: PMC6487713 DOI: 10.1002/1878-0261.12488] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/07/2019] [Accepted: 04/04/2019] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer death worldwide. HCC treatment is hindered by the frequent emergence of chemoresistance to the multikinase inhibitor sorafenib, which has been related to the presence of cancer stem cells (CSCs) that self‐renew and often escape therapy. The key metabolic sensor AMP‐activated kinase (AMPK) has recently been recognized as a tumour growth regulator. In this study, we aimed to elucidate the role of AMPK in the development of a stem cell phenotype in HCC cells. To this end, we enriched the CSC population in HCC cell lines that showed increased expression of drug resistance (ALDH1A1, ABCB1A) and stem cell (CD133, Nanog, Oct4, alpha fetoprotein) markers and demonstrated their stemness phenotype. These cells were refractory to sorafenib‐induced cell death. We report that sorafenib‐resistant cells had lower levels of total and phosphorylated AMPK as well as its downstream substrate, ACC, compared with the parental cells. Interestingly, AMPK knockdown with siRNA or inhibition with dorsomorphin increased the expression of stem cell markers in parental cells and blocked sorafenib‐induced cell death. Conversely, the upregulation of AMPK, either by transfection or by pharmacological activation with A‐769662, decreased the expression of ALDH1A1, ABCB1A, CD133, Nanog, Oct4, and alpha fetoprotein, and restored sensitivity to sorafenib. Analysis of the underlying mechanism points to hypoxia‐inducible factor HIF‐1α as a regulator of stemness. In vivo studies in a xenograft mouse model demonstrated that stem‐like cells have greater tumourigenic capacity. AMPK activation reduced xenograft tumour growth and decreased the expression of stem cell markers. Taken together, these results indicate that AMPK may serve as a novel target to overcome chemoresistance in HCC.
Collapse
Affiliation(s)
- Alicia Bort
- Department of Systems Biology, School of Medicine, University of Alcala, Alcalá de Henares, Madrid, Spain
| | - Belén G Sánchez
- Department of Systems Biology, School of Medicine, University of Alcala, Alcalá de Henares, Madrid, Spain
| | - Pedro A Mateos-Gómez
- Department of Systems Biology, School of Medicine, University of Alcala, Alcalá de Henares, Madrid, Spain
| | - Diana Vara-Ciruelos
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, UK
| | - Nieves Rodríguez-Henche
- Department of Systems Biology, School of Medicine, University of Alcala, Alcalá de Henares, Madrid, Spain
| | - Inés Díaz-Laviada
- Department of Systems Biology, School of Medicine, University of Alcala, Alcalá de Henares, Madrid, Spain.,Chemical Research Institute 'Andrés M. del Río' (IQAR), Alcalá University, Alcalá de Henares, Madrid, Spain
| |
Collapse
|
37
|
Deng J, Peng M, Wang Z, Zhou S, Xiao D, Deng J, Yang X, Peng J, Yang X. Novel application of metformin combined with targeted drugs on anticancer treatment. Cancer Sci 2018; 110:23-30. [PMID: 30358009 PMCID: PMC6317954 DOI: 10.1111/cas.13849] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/17/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022] Open
Abstract
The success of targeted drug therapies for treating cancer patients has attracted broad attention both in the academic community and social society. However, rapidly developed acquired resistance is becoming a newly recognized major challenge to the continuing efficiency of these therapies. Metformin is a well‐known natural compound with low toxicity derived from the plant French lilac. Our previous work has highlighted research progress of the combination of clinically applied chemotherapies and metformin by different mechanisms. We have also launched a study to combine metformin with the small molecule targeted drug gefitinib to treat bladder cancer using intravesical administration. Thus, in this minireview, we summarize recent achievements combining metformin with various targeted therapies. This work directs the potential clinical future by selecting available cancer patients and providing precise medicine by the combination of metformin and targeted drugs to overcome resistance and enhance therapeutic efficacies.
Collapse
Affiliation(s)
- Jun Deng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Mei Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiren Wang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Sichun Zhou
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Di Xiao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Jiating Deng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Xue Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingyuan Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
38
|
Chung YK, Hwang S, Song GW, Lee YJ, Kim KH, Ahn CS, Moon DB, Ha TY, Jung DH, Park GC, Ryoo BY, Lee SG. Absence of antitumor effects of metformin in sorafenib-treated patients with hepatocellular carcinoma recurrence after hepatic resection and liver transplantation. Ann Hepatobiliary Pancreat Surg 2018; 22:297-304. [PMID: 30588519 PMCID: PMC6295365 DOI: 10.14701/ahbps.2018.22.4.297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 06/10/2018] [Accepted: 06/14/2018] [Indexed: 01/27/2023] Open
Abstract
Backgrounds/Aims Hepatocellular carcinoma (HCC) recurrence following hepatic resection (HR) and liver transplantation (LT) remains a great concern. We assessed the antitumor effects of metformin in patients treated with sorafenib for HCC recurrence after HR or LT. Methods The two clinical retrospective studies involved metformin therapy of 304 HR patients and 74 LT recipients who were treated with sorafenib. Results In the study involving patients who had undergone HR, death occurred in 245 of the 304 patients (80.6%) during a median follow-up of 10.2 months after sorafenib administration. The metformin HR group (group 1; n=40) showed no prognostic difference in progression-free and overall survival rates compared with the all-HR control group (group 3; n=241) and propensity score-matched HR control group (group 4; n=80). In the clinical study of recipients exposed to LT, death occurred in 62 of the 74 patients (83.8%) during a median follow-up of 13.6 months (range: 3–76 months) after sorafenib administration. The metformin LT group (group 5; n=14) showed no prognostic difference in progression-free and overall survival rates compared with the all-LT control group (group 7; n=43) and propensity score-matched LT control group (group 8; n=28). Conclusions Our clinical studies demonstrated absence of synergistic antitumor effects of metformin. Further high-volume studies are necessary to assess the role of metformin in patients treated with sorafenib for advanced HCC.
Collapse
Affiliation(s)
- Yong-Kyu Chung
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shin Hwang
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gi-Won Song
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young-Joo Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ki-Hun Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chul-Soo Ahn
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Deok-Bog Moon
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Tae-Yong Ha
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dong-Hwan Jung
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gil-Chun Park
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung-Gyu Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
39
|
Yin X, Wei Z, Song C, Tang C, Xu W, Wang Y, Xie J, Lin Z, Han W. Metformin sensitizes hypoxia-induced gefitinib treatment resistance of HNSCC via cell cycle regulation and EMT reversal. Cancer Manag Res 2018; 10:5785-5798. [PMID: 30510448 PMCID: PMC6250113 DOI: 10.2147/cmar.s177473] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objectives The objectives of this study were to explore the mechanisms of metformin sensitization to hypoxia-induced gefitinib treatment in resistant head and neck squamous cell carcinoma (HNSCC) and evaluate the effects of this combined treatment strategy. Methods The effects of gefitinib treatment on HNSCC were measured under normoxic and hypoxic conditions. The relationship between hypoxia and cell cycle and epithelial-mesenchymal transition (EMT) in tumor cells were analyzed. Palbociclib and LY294002 were used in combination with gefitinib to evaluate the effects on HNSCC cell cytotoxicity during hypoxia. Finally, metformin was used to evaluate the sensitizing effects of gefitinib treatment on HNSCC in vivo and in vitro. Results Cell viability and apoptosis assays demonstrated a significant difference in HNSCC cells treated with gefitinib between the normoxia and hypoxia groups. Hypoxia induced the expression of cyclin D1, decreased the percentage of cells in G1, and promoted the EMT of tumor cells. Both palbociclib and LY294002 enhanced gefitinib-induced cytotoxicity of HNSCC cells under hypoxic conditions. Encouragingly, metformin sensitized HNSCC to gefitinib treatment in vivo and in vitro. Conclusion Hypoxia promotes G1-S cell cycle progression and EMT in HNSCC, resulting in gefitinib treatment resistance. Metformin sensitizes HNSCC to gefitinib treatment, which might serve as a novel combined treatment strategy.
Collapse
Affiliation(s)
- Xiteng Yin
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, .,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, ,
| | - Zheng Wei
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, .,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, ,
| | - Chuanhui Song
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, .,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, ,
| | - Chuanchao Tang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, .,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, ,
| | - Wenguang Xu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, .,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, ,
| | - Yufeng Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, .,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, ,
| | - Junqi Xie
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, .,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, ,
| | - Zitong Lin
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, , .,Department of Dentomaxillofacial Radiology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China,
| | - Wei Han
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, .,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China, ,
| |
Collapse
|
40
|
Méndez-Blanco C, Fondevila F, García-Palomo A, González-Gallego J, Mauriz JL. Sorafenib resistance in hepatocarcinoma: role of hypoxia-inducible factors. Exp Mol Med 2018; 50:1-9. [PMID: 30315182 PMCID: PMC6185986 DOI: 10.1038/s12276-018-0159-1] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/05/2018] [Accepted: 06/18/2018] [Indexed: 12/14/2022] Open
Abstract
Sorafenib, a multikinase inhibitor with antiproliferative, antiangiogenic, and proapoptotic properties, constitutes the only effective first-line drug approved for the treatment of advanced hepatocellular carcinoma (HCC). Despite its capacity to increase survival in HCC patients, its success is quite low in the long term owing to the development of resistant cells through several mechanisms. Among these mechanisms, the antiangiogenic effects of sustained sorafenib treatment induce a reduction of microvessel density, promoting intratumoral hypoxia and hypoxia-inducible factors (HIFs)-mediated cellular responses that favor the selection of resistant cells adapted to the hypoxic microenvironment. Clinical data have demonstrated that overexpressed HIF-1α and HIF-2α in HCC patients are reliable markers of a poor prognosis. Thus, the combination of current sorafenib treatment with gene therapy or inhibitors against HIFs have been documented as promising approaches to overcome sorafenib resistance both in vitro and in vivo. Because the depletion of one HIF-α subunit elevates the expression of the other HIF-α isoform through a compensatory loop, targeting both HIF-1α and HIF-2α would be a more interesting strategy than therapies that discriminate among HIF-α isoforms. In conclusion, there is a marked correlation between the hypoxic microenvironment and sorafenib resistance, suggesting that targeting HIFs is a promising way to increase the efficiency of treatment. Targeting hypoxia-inducible factors (HIFs), regulatory proteins induced by low oxygen levels, could increase the effectiveness of sorafenib, the only systemic therapy approved for advanced liver cancer. Long-term treatment with sorafenib starves tumors of oxygen, which can lead to the proliferation of cancer cells that are able to survive low oxygen levels. HIFs regulate genes involved in this adaptation and HIF levels are increased in sorafenib-resistant cells. José Mauriz at the University of León, Spain, and colleagues review recent studies on the effects of HIF inhibition on sorafenib efficacy. They conclude that HIF-1α and HIF-2α are predictive markers of sorafenib resistance and that using inhibitors of both these factors as an add-on therapy could improve patient survival. This strategy may be applicable to other types of cancer in which reduced oxygen conditions lead to drug resistance.
Collapse
Affiliation(s)
- Carolina Méndez-Blanco
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Flavia Fondevila
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Andrés García-Palomo
- Institute of Biomedicine, University of León, León, Spain.,Service of Oncology, Complejo Asistencial Universitario de León, León, Spain
| | - Javier González-Gallego
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - José L Mauriz
- Institute of Biomedicine, University of León, León, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain.
| |
Collapse
|
41
|
Extracellular matrix collagen I promotes the tumor progression of residual hepatocellular carcinoma after heat treatment. BMC Cancer 2018; 18:901. [PMID: 30227844 PMCID: PMC6145107 DOI: 10.1186/s12885-018-4820-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 09/13/2018] [Indexed: 12/29/2022] Open
Abstract
Background Accelerated malignant behaviors induced by insufficient thermal ablation have been increasingly reported, however, the exact mechanisms are still unclear. Here, we investigated the importance of the extracellular matrix (ECM) in modulating the progression of residual hepatocellular carcinoma (HCC) after heat treatment. Methods Heat-exposed residual HCC cells were cultured in different ECM gels. We used basement membrane gel (Matrigel) to simulate the normal microenvironment and collagen I to model the pathological stromal ECM. The alterations of morphology and parameters of proliferation, epithelial-mesenchymal transition (EMT) and stemness were analyzed in vitro and in vivo. Results Increased collagen I deposition was observed at the periablational zone after incomplete RFA of HCC in a xenograft model. The markers of cell proliferation, EMT, motility and progenitor-like traits of heat-exposed residual HCC cells were significantly induced by collagen I as compared to Matrigel (p values all < 0.05). Importantly, collagen I induced the activation of ERK phosphorylation in heat-exposed residual HCC cells. ERK1/2 inhibitor reversed the collagen I-promoted ERK phosphorylation, cell proliferative, protrusive and spindle-like appearance of heat-treated residual HCC cells in vitro. Moreover, collagen I promoted the in vivo tumor progression of heat-exposed residual HCC cells, and sorafenib markedly reversed the collagen I-mediated protumor effects. Conclusions Our findings demonstrate that collagen I could enhance the aggressive progression of residual HCC cells after suboptimal heat treatment and sorafenib may be a treatment approach to thwart this process. Electronic supplementary material The online version of this article (10.1186/s12885-018-4820-9) contains supplementary material, which is available to authorized users.
Collapse
|
42
|
Feng Y, Guo X, Huang X, Wu M, Li X, Wu S, Luo X. Metformin reverses stem cell‑like HepG2 sphere formation and resistance to sorafenib by attenuating epithelial‑mesenchymal transformation. Mol Med Rep 2018; 18:3866-3872. [PMID: 30106125 PMCID: PMC6131649 DOI: 10.3892/mmr.2018.9348] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/05/2018] [Indexed: 12/19/2022] Open
Abstract
Cancer stem cells (CSCs) have been reported to be associated with the recurrence and drug resistance of liver cancer. In the present study, stem cell-like HepG2 cell spheres were enriched using stem cell conditioned culture medium. As expected, stem-like HepG2 cell spheres exhibited increased resistance to sorafenib. Metformin, a common drug used to treat type 2 diabetes mellitus, reduced the diameters and numbers of stem-like HepG2 spheres, and increased their sensitivity to sorafenib. Western blotting confirmed that low doses of metformin reversed the epithelial-mesenchymal transformation (EMT) process of HepG2 spheres. These results suggested that metformin enhanced sensitivity to sorafenib, which was probably through reversal of the EMT process of sphere-forming cells and by reducing the formation of CSCs.
Collapse
Affiliation(s)
- Yan Feng
- Research Department, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xing Guo
- Research Department, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xinping Huang
- Research Department, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Manya Wu
- Research Department, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xin Li
- Research Department, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Shushu Wu
- Research Department, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoling Luo
- Research Department, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
43
|
Saran U, Guarino M, Rodríguez S, Simillion C, Montani M, Foti M, Humar B, St-Pierre MV, Dufour JF. Anti-tumoral effects of exercise on hepatocellular carcinoma growth. Hepatol Commun 2018; 2:607-620. [PMID: 29761175 PMCID: PMC5944574 DOI: 10.1002/hep4.1159] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/11/2018] [Indexed: 12/19/2022] Open
Abstract
Regular physical exercise has many beneficial effects, including antitumor properties, and is associated with a reduced risk of developing hepatocellular carcinoma (HCC). Less is known about the impact of exercise on HCC growth and progression. Here, we investigated the effects of exercise on HCC progression and assessed whether any beneficial effects would be evident under sorafenib treatment and could be mimicked by metformin. American Cancer Institute rats with orthotopic syngeneic HCC derived from Morris Hepatoma‐3924A cells were randomly assigned to exercise (Exe) and sedentary groups, or sorafenib±Exe groups or sorafenib±metformin groups. The Exe groups ran on a motorized treadmill for 60 minutes/day, 5 days/week for 4 weeks. Tumor viable area was decreased by exercise, while cell proliferation and vascular density were reduced. Exercise increased the expression of phosphatase and tensin homolog deleted from chromosome 10 and increased the phosphorylation of adenosine monophosphate‐activated protein kinase, while the phosphorylation of protein kinase B, S6 ribosomal protein, and signal transducer and activator of transcription 3 were decreased. Transcriptomic analysis suggested major effects of exercise were on nontumoral liver rather than tumor tissue. Exercise demonstrated similar effects when combined with sorafenib. Moreover, similar effects were observed in the group treated with sorafenib+metformin, revealing an exercise‐mimicking effect of metformin. Conclusion: Exercise attenuates HCC progression associated with alterations in key signaling pathways, cellular proliferation, tumor vascularization, and necrosis. These beneficial effects are maintained when combined with sorafenib and can be mimicked by metformin. (Hepatology Communications 2018;2:607‐620)
Collapse
Affiliation(s)
- Uttara Saran
- Hepatology Section, Department for BioMedical Research University of Bern Bern Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital Bern Bern Switzerland
| | - Maria Guarino
- Hepatology Section, Department for BioMedical Research University of Bern Bern Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital Bern Bern Switzerland.,Gastroenterology Section, Department of Clinical Medicine and Surgery University of Naples "Federico II," Naples Italy
| | - Sarai Rodríguez
- Hepatology Section, Department for BioMedical Research University of Bern Bern Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital Bern Bern Switzerland
| | - Cedric Simillion
- Institute for Bioinformatics University of Bern Bern Switzerland
| | | | - Michelangelo Foti
- Department of Cell Physiology and Metabolism University of Geneva Geneva Switzerland
| | - Bostjan Humar
- Department of Visceral and Transplantation Surgery University Hospital Zürich Zürich Switzerland
| | - Marie V St-Pierre
- Hepatology Section, Department for BioMedical Research University of Bern Bern Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital Bern Bern Switzerland
| | - Jean-François Dufour
- Hepatology Section, Department for BioMedical Research University of Bern Bern Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital Bern Bern Switzerland
| |
Collapse
|
44
|
Saini N, Yang X. Metformin as an anti-cancer agent: actions and mechanisms targeting cancer stem cells. Acta Biochim Biophys Sin (Shanghai) 2018; 50:133-143. [PMID: 29342230 DOI: 10.1093/abbs/gmx106] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022] Open
Abstract
Metformin, a first line medication for type II diabetes, initially entered the spotlight as a promising anti-cancer agent due to epidemiologic reports that found reduced cancer risk and improved clinical outcomes in diabetic patients taking metformin. To uncover the anti-cancer mechanisms of metformin, preclinical studies determined that metformin impairs cellular metabolism and suppresses oncogenic signaling pathways, including receptor tyrosine kinase, PI3K/Akt, and mTOR pathways. Recently, the anti-cancer potential of metformin has gained increasing interest due to its inhibitory effects on cancer stem cells (CSCs), which are associated with tumor metastasis, drug resistance, and relapse. Studies using various cancer models, including breast, pancreatic, prostate, and colon, have demonstrated the potency of metformin in attenuating CSCs through the targeting of specific pathways involved in cell differentiation, renewal, metastasis, and metabolism. In this review, we provide a comprehensive overview of the anti-cancer actions and mechanisms of metformin, including the regulation of CSCs and related pathways. We also discuss the potential anti-cancer applications of metformin as mono- or combination therapies.
Collapse
Affiliation(s)
- Nipun Saini
- Julius L. Chambers Biomedical/Biotechnology Research Institute, Department of Biological and Biomedical Sciences, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - Xiaohe Yang
- Julius L. Chambers Biomedical/Biotechnology Research Institute, Department of Biological and Biomedical Sciences, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC 28081, USA
| |
Collapse
|
45
|
Cao MQ, You AB, Zhu XD, Zhang W, Zhang YY, Zhang SZ, Zhang KW, Cai H, Shi WK, Li XL, Li KS, Gao DM, Ma DN, Ye BG, Wang CH, Qin CD, Sun HC, Zhang T, Tang ZY. miR-182-5p promotes hepatocellular carcinoma progression by repressing FOXO3a. J Hematol Oncol 2018; 11:12. [PMID: 29361949 PMCID: PMC5782375 DOI: 10.1186/s13045-018-0555-y] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 01/15/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND High frequency of recurrence is the major cause of the poor outcomes for patients with hepatocellular carcinoma (HCC). microRNA (miR)-182-5p emerged as a high-priority miRNA in HCC and was found to be related to HCC metastasis. Whether the expression of miR-182-5p in tumor tissue correlated with early recurrence in HCC patients underwent curative surgery was unknown. METHODS Real-time PCR (RT-PCR) and in situ hybridization (ISH) were conducted to assess the expression of miR-182-5p in HCC cells and tissues. Cell Counting Kit-8 (CCK-8), transwell assays were performed to detected cells proliferation and migration ability. Flow cytometry assays were used to detect cell apoptosis rate, and xenograft model was employed to study miR-182-5p in HCC growth and lung metastasis. The target of miR-182-5p was validated with a dual-luciferase reporter assay and western blotting. Immunohistochemistry, immumoblotting, and immunoprecipitation were performed to test relative protein expression. RESULTS We showed that high expression of miR-182-5p in tumor tissues correlated with poor prognosis as well as early recurrence in HCC patients underwent curative surgery. miR-182-5p enhanced motility and invasive ability of HCC cells both in vitro and in vivo. miR-182-5p directly targets 3'-UTR of FOXO3a and repressed FOXO3a expression, activating AKT/FOXO3a pathway to promote HCC proliferation. Notably, miR-182-5p activated Wnt/β-catenin signaling by inhibiting the degradation of β-catenin and enhancing the interaction between β-catenin and TCF4 which was mediated by repressed FOXO3a. CONCLUSIONS Consistently, miR-182-5p can be a potential predictor of early recurrence for HCC patients underwent curative surgery, and FOXO3a plays a key mediator in miR-182-5p induced HCC progression.
Collapse
Affiliation(s)
- Man-Qing Cao
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - A-Bin You
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Haidian District, Beijing, 100142, China
| | - Xiao-Dong Zhu
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Wei Zhang
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Yuan-Yuan Zhang
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Shi-Zhe Zhang
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Ke-Wei Zhang
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Hao Cai
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Wen-Kai Shi
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Xiao-Long Li
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Kang-Shuai Li
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Dong-Mei Gao
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - De-Ning Ma
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Zhejiang, Hangzhou, 310022, China
| | - Bo-Gen Ye
- Department of Organ Transplantation, Changhai Hospital, The Second Military Medical University, Shanghai, 200433, China
| | - Cheng-Hao Wang
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, 200032, China
| | - Cheng-Dong Qin
- Department of Breast Cancer Surgery, Zhejiang Cancer Hospital, Zhejiang, Hangzhou, 310022, China
| | - Hui-Chuan Sun
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Ti Zhang
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
| | - Zhao-You Tang
- Department of Hepatobiliary Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
46
|
Metformin ameliorates arsenic trioxide hepatotoxicity via inhibiting mitochondrial complex I. Cell Death Dis 2017; 8:e3159. [PMID: 29095437 PMCID: PMC5775401 DOI: 10.1038/cddis.2017.482] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/19/2017] [Accepted: 08/22/2017] [Indexed: 12/11/2022]
Abstract
Arsenic trioxide (ATO) is a well-accepted chemotherapy agent in managing promyelocytic leukemia. ATO often causes severe health hazards such as hepatotoxicity, dermatosis, neurotoxicity, nephrotoxicity and cardiotoxicity. The production of reactive oxygen species, (ROS) play a significant role in ATO-induced hepatotoxicity. The oral hypoglycemic drug, metformin, is considered to be a potential novel agent for chemoprevention in the treatment of cancer. Moreover, metformin has also been shown to have hepatoprotective effects. In the present study, we demonstrated that metformin protected normal hepatocytes from ATO-induced apoptotic cell death in vitro and in vivo. Gene expression screening revealed that glucose metabolism might be related to the metformin-induced protective effect on ATO-treated AML12 cells. The metformin-promoted or induced glycolysis was not responsible for the protection of AML12 cells from ATO-induced apoptotic cell death. Instead, metformin increased the intracellular NADH/NAD+ ratio by inhibiting mitochondrial respiratory chain complex I, further decreasing the intracellular ROS induced by ATO. Treatment with low glucose or rotenone, a mitochondrial respiratory chain complex I inhibitor, also protected AML12 cells from ATO-induced apoptotic cell death. We show for the first time that metformin protects the hepatocyte from ATO by regulating the mitochondrial function. With its properties of chemoprevention, chemosensitization and the amelioration of liver damage, metformin has great prospects for clinical application other than type 2 diabetes mellitus (T2DM).
Collapse
|
47
|
Capsaicin exerts synergistic antitumor effect with sorafenib in hepatocellular carcinoma cells through AMPK activation. Oncotarget 2017; 8:87684-87698. [PMID: 29152112 PMCID: PMC5675664 DOI: 10.18632/oncotarget.21196] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/25/2017] [Indexed: 12/22/2022] Open
Abstract
In this study, we investigated the antitumoral effects of combined treatment using sorafenib and capsaicin in hepatocellular carcinoma (HCC) cells. Here we showed that the combination of the two drugs had a much stronger inhibitory effect on both HepG2 and Huh-7 human HCC cells growth than either drug alone. The isobolograms demonstrated that the combinations investigated in this study produced a synergistic interaction. In the combination treatment using capsaicin and sorafenib, increased apoptosis, followed by the activation of caspase-9 and PARP, was observed. In addition, the present study demonstrated that sorafenib treatment induces activation of Akt, probably as a mechanism of resistance, whereas capsaicin inhibits Akt providing a possible pathway whereby capsaicin sensitizes to sorafenib in HCC cells. Moreover, capsaicin singly and the combination of capsaicin and sorafenib induce AMPK activation and Acetyl CoA carboxylase phosphorylation in HCC cells. Knocking down of AMPK by selective siRNA abrogates capsaicin-induced Akt inhibition, suggesting the involvement of AMPK in the antiproliferative effect. In vivo experiments further showed that that the anti-tumor effect of sorafenib was enhanced by its combination with 2.5 mg/Kg of capsaicin. Overall, these results show that combined treatment with capsaicin and sorafenib might improve sorafenib sensitivity and therefore it represents a promising and attractive strategy for the treatment of HCC.
Collapse
|
48
|
Kast RE, Skuli N, Karpel-Massler G, Frosina G, Ryken T, Halatsch ME. Blocking epithelial-to-mesenchymal transition in glioblastoma with a sextet of repurposed drugs: the EIS regimen. Oncotarget 2017; 8:60727-60749. [PMID: 28977822 PMCID: PMC5617382 DOI: 10.18632/oncotarget.18337] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/12/2017] [Indexed: 12/11/2022] Open
Abstract
This paper outlines a treatment protocol to run alongside of standard current treatment of glioblastoma- resection, temozolomide and radiation. The epithelial to mesenchymal transition (EMT) inhibiting sextet, EIS Regimen, uses the ancillary attributes of six older medicines to impede EMT during glioblastoma. EMT is an actively motile, therapy-resisting, low proliferation, transient state that is an integral feature of cancers’ lethality generally and of glioblastoma specifically. It is believed to be during the EMT state that glioblastoma’s centrifugal migration occurs. EMT is also a feature of untreated glioblastoma but is enhanced by chemotherapy, by radiation and by surgical trauma. EIS Regimen uses the antifungal drug itraconazole to block Hedgehog signaling, the antidiabetes drug metformin to block AMP kinase (AMPK), the analgesic drug naproxen to block Rac1, the anti-fibrosis drug pirfenidone to block transforming growth factor-beta (TGF-beta), the psychiatric drug quetiapine to block receptor activator NFkB ligand (RANKL) and the antibiotic rifampin to block Wnt- all by their previously established ancillary attributes. All these systems have been identified as triggers of EMT and worthy targets to inhibit. The EIS Regimen drugs have a good safety profile when used individually. They are not expected to have any new side effects when combined. Further studies of the EIS Regimen are needed.
Collapse
Affiliation(s)
| | - Nicolas Skuli
- INSERM, Centre de Recherches en Cancérologie de Toulouse, CRCT, Inserm/Université Toulouse III, Paul Sabatier, Hubert Curien, Toulouse, France
| | - Georg Karpel-Massler
- Department of Neurosurgery, Ulm University Hospital, Albert-Einstein-Allee, Ulm, Germany
| | - Guido Frosina
- Mutagenesis & Cancer Prevention Unit, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Largo Rosanna Benzi, Genoa, Italy
| | - Timothy Ryken
- Department of Neurosurgery, University of Kansas, Lawrence, KS, USA
| | - Marc-Eric Halatsch
- Department of Neurosurgery, Ulm University Hospital, Albert-Einstein-Allee, Ulm, Germany
| |
Collapse
|
49
|
Zhang Q, Kong J, Dong S, Xu W, Sun W. Metformin exhibits the anti-proliferation and anti-invasion effects in hepatocellular carcinoma cells after insufficient radiofrequency ablation. Cancer Cell Int 2017; 17:48. [PMID: 28450808 PMCID: PMC5404300 DOI: 10.1186/s12935-017-0418-6] [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: 12/17/2016] [Accepted: 04/18/2017] [Indexed: 02/06/2023] Open
Abstract
Background The mechanisms and prevention of progression of hepatocellular carcinoma (HCC) after insufficient radiofrequency ablation (RFA) has been preliminarily investigated, therefore, new strategy needs to be investigated to prevent the process. Whether metformin could be used to inhibit the growth of HCC after insufficient RFA and further prevent the progression of residual HCC remains unclearly. Methods MTT assay, colony formation assay and transwell assay were used to observe the cell viability, migration and invasion. Western blot and immunohistochemistry methods were used to observe the expression of proteins. Xenograft model was used to evaluate the growth of HCC cells in vivo. Results Metformin inhibited the enhanced proliferation, migration and invasion of HepG2 and SMMC7721 cells after insufficient RFA (named as HepG2-H and SMMC7721-H). Metformin deregulated the expression of p-Akt in HepG2 and SMMC7721 cells after insufficient RFA through AMPK/PTEN pathway. HepG2-H cells also exhibited larger tumor size in vivo. Higher expression of Ki-67 and CD31 and lower expression of E-cadherin were observed in HepG2-H tumors. Metformin blocked the enhanced growth of HepG2 cells in vivo after insufficient RFA. Metformin had no apparent toxicity on nude mice. Conclusions Metfromin inhibited the growth of HCC cells after insufficient RFA, and may be used to prevent the progression of HCC after RFA. Electronic supplementary material The online version of this article (doi:10.1186/s12935-017-0418-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qingyun Zhang
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100043 China.,Department of General Surgery, Affiliated Hospital of Chengde Medical University, Hebei, 067000 China
| | - Jian Kong
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100043 China
| | - Shuying Dong
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100043 China
| | - Wenlei Xu
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100043 China
| | - Wenbing Sun
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100043 China
| |
Collapse
|
50
|
Zheng Y, Jiang L, Hu Y, Xiao C, Xu N, Zhou J, Zhou X. Metallothionein 1H (MT1H) functions as a tumor suppressor in hepatocellular carcinoma through regulating Wnt/β-catenin signaling pathway. BMC Cancer 2017; 17:161. [PMID: 28241806 PMCID: PMC5330125 DOI: 10.1186/s12885-017-3139-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 02/15/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Metallothionein 1H (MT1H) expression level is downregulated in several kinds of tumors, including hepatocellular cancer (HCC). However, its biological functions and underlying mechanisms in HCC is largely unknown. The current study aimed to demonstrate the expression status, biological roles and potential mechanisms of MT1H in HCC. METHODS We investigated the expression level of MT1H in the Cancer Genome Atlas (TCGA) dataset and a panel of 12 paired tumor/non-tumor tissues. In vitro, gain-of-function experiments were performed to examine the role of MT1H on HCC cell proliferation, invasion, and migration. Using bioinformatics assay, reporter assays, quantitative real-time PCR, and western blotting, we explored the possible mechanisms underlying the role of MT1H in HCC cells. In vivo nude mice experiments were performed to assess the anti-proliferative role of MT1H in HCC. RESULTS Downregulation of MT1H was observed in TCGA dataset and a panel of 12 paired tumor/non-tumor tissues. Ectopic overexpression of MT1H in HepG2 and Hep3B cells inhibited cell proliferation, invasion, and migration. Gene Set Enrichment Analysis (GSEA) showed that MT1H might involve in regulation of Wnt/β-catenin pathway. Top/Fop reporter assay confirmed that MT1H had an effect on Wnt/β-catenin signaling. Real-time PCR showed MT1H expression decreased the expression of Wnt/β-catenin target genes. Western blotting assay showed that overexpression of MT1H inhibited the nuclear translocation of β-catenin and that the Akt/GSK-3β axis mediated the modulatory role of MT1H on Wnt/β-catenin signaling in HCC. In vivo nude mice experiments demonstrated that MT1H suppressed the proliferation of HCC cells. Taken together, MT1H suppressed the proliferation, invasion and migration of HCC cells via regulating Wnt/β-catenin signaling pathway. CONCLUSIONS This study demonstrated that through inhibiting Wnt/β-catenin pathway, MT1H suppresses the proliferation and invasion of HCC cells. MT1H may be a potential target for HCC therapy.
Collapse
Affiliation(s)
- Yulong Zheng
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lihua Jiang
- Department of Neurology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, 31006, China
| | - Yongxian Hu
- Department of Hematology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Cheng Xiao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jianying Zhou
- Department of Respiratory Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| | - Xinhui Zhou
- Department of Gynecology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|