1
|
Zhao J, Lei L, Dai W, Jiang A, Jin Q, Tang Z. Simultaneous inhibition of heat shock proteins and autophagy enhances radiofrequency ablation of hepatocellular carcinoma. Biomater Sci 2024. [PMID: 39429155 DOI: 10.1039/d4bm01190b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Radiofrequency ablation (RFA) is a commonly used minimally invasive treatment for hepatocellular carcinoma (HCC). However, incomplete radiofrequency ablation (iRFA) promotes tumor progression and metastasis. There is an urgent need to develop innovative strategies to enhance the efficacy of iRFA. The upregulation of heat shock proteins (HSPs) and activation of protective autophagy in tumor cells upon exposure to sublethal heat enhance the thermotolerance, thereby promoting tumor cell survival. Here, 3-methyladenine (3-MA) and lonidamine (LND) co-encapsulated liposomes (Lip@LND/3-MA) are designed to enhance the efficacy of iRFA by simultaneous inhibition of glycolysis and autophagy. On one hand, LND inhibits hexokinase, a key enzyme in glycolysis, and thus reduces ATP production and consequently suppresses the expression of HSPs. On the other hand, 3-MA, as an autophagy inhibitor, can inhibit protective autophagy after iRFA. Lip@LND/3-MA is confirmed to suppress the expression of HSPs and reduce the autophagy level during RFA. Therefore, the thermotolerance of tumor cells is significantly weakened, leading to remarkably enhanced therapeutic efficacy of iRFA. It is believed that simultaneous inhibition of HSPs and autophagy is a promising therapeutic strategy in clinical practice of RFA.
Collapse
Affiliation(s)
- Jinchao Zhao
- Department of General Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China.
| | - Lei Lei
- Department of General Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China.
| | - Wenbin Dai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China.
| | - Angfeng Jiang
- Department of General Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China.
| | - Qiao Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China.
| | - Zhe Tang
- Department of General Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China.
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| |
Collapse
|
2
|
Wu J, Zhou Z, Huang Y, Deng X, Zheng S, He S, Huang G, Hu B, Shi M, Liao W, Huang N. Radiofrequency ablation: mechanisms and clinical applications. MedComm (Beijing) 2024; 5:e746. [PMID: 39359691 PMCID: PMC11445673 DOI: 10.1002/mco2.746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 10/04/2024] Open
Abstract
Radiofrequency ablation (RFA), a form of thermal ablation, employs localized heat to induce protein denaturation in tissue cells, resulting in cell death. It has emerged as a viable treatment option for patients who are ineligible for surgery in various diseases, particularly liver cancer and other tumor-related conditions. In addition to directly eliminating tumor cells, RFA also induces alterations in the infiltrating cells within the tumor microenvironment (TME), which can significantly impact treatment outcomes. Moreover, incomplete RFA (iRFA) may lead to tumor recurrence and metastasis. The current challenge is to enhance the efficacy of RFA by elucidating its underlying mechanisms. This review discusses the clinical applications of RFA in treating various diseases and the mechanisms that contribute to the survival and invasion of tumor cells following iRFA, including the roles of heat shock proteins, hypoxia, and autophagy. Additionally, we analyze the changes occurring in infiltrating cells within the TME after iRFA. Finally, we provide a comprehensive summary of clinical trials involving RFA in conjunction with other treatment modalities in the field of cancer therapy, aiming to offer novel insights and references for improving the effectiveness of RFA.
Collapse
Affiliation(s)
- Jianhua Wu
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Zhiyuan Zhou
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yuanwen Huang
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xinyue Deng
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Siting Zheng
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Shangwen He
- Department of Respiratory and Critical Care MedicineChronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical UniversityGuangzhouGuangdongChina
| | - Genjie Huang
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Binghui Hu
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Min Shi
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Wangjun Liao
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Na Huang
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
3
|
Li X, Lv Z, Zhou P, Zhang S, Jiang C. Sox9: A potential regulator of cancer stem cells in osteosarcoma. Open Med (Wars) 2024; 19:20240995. [PMID: 38978960 PMCID: PMC11229887 DOI: 10.1515/med-2024-0995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/13/2024] [Accepted: 06/14/2024] [Indexed: 07/10/2024] Open
Abstract
Osteosarcoma is a highly aggressive bone tumor primarily affecting children and adolescents. Despite advancements in treatment modalities, the prognosis for osteosarcoma patients remains poor, emphasizing the need for a deeper understanding of its underlying mechanisms. In recent years, the concept of cancer stem cells (CSCs) has emerged as a crucial factor in tumor initiation, progression, and therapy resistance. These specialized subpopulations of cells possess self-renewal capacity, tumorigenic potential, and contribute to tumor heterogeneity. Sox9, a transcription factor known for its critical role in embryonic development and tissue homeostasis, has been implicated in various malignancies, including osteosarcoma. This review aims to summarize the current knowledge regarding the role of Sox9 in CSCs in osteosarcoma and its potential implications as a prognosis and therapeutic target.
Collapse
Affiliation(s)
- Xiucheng Li
- Department of Orthopaedics, Shaoxing People’ Hospital, Shaoxing, China
| | - Zuo Lv
- Department of Orthopaedics, Shaoxing People’ Hospital, Shaoxing, China
| | - Ping Zhou
- Department of Orthopaedics, Shaoxing People’ Hospital, Shaoxing, China
| | - SongOu Zhang
- Department of Orthopaedics, Shaoxing People’ Hospital, Shaoxing, China
- School of Medicine, Ningbo University, Ningbo, China
| | - Chao Jiang
- Department of Orthopaedics, Shaoxing People’ Hospital, Shaoxing, China
| |
Collapse
|
4
|
Kuo CY, Tsai CH, Wu JK, Cheng SP. Sublethal thermal stress promotes migration and invasion of thyroid cancer cells. PLoS One 2024; 19:e0298903. [PMID: 38394093 PMCID: PMC10889624 DOI: 10.1371/journal.pone.0298903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE Radiofrequency ablation is a viable option in the treatment of benign thyroid nodules. Some reports suggest that thermal ablation may also be safe for the management of low-risk thyroid cancer. In this study, we applied transient heat treatment to thyroid cancer cells to mimic clinical scenarios in which insufficient ablation leads to incomplete eradication of thyroid cancer. METHODS Differentiated thyroid cancer cell lines B-CPAP, TPC-1, and FTC-133 were subjected to heat treatment at different temperatures for 10 min. Effects on cell growth, clonogenicity, wound healing assay, and Transwell invasion were determined. RESULTS Heat treatment at 45°C or higher reduced cell growth, whereas viability of thyroid cancer cells was not changed after heat treatment at 37, 40, or 42°C. Heat treatment at 40°C increased the number of colony formations by 16% to 39%. Additionally, transient heat treatment at 40°C resulted in a 1.75-fold to 2.56-fold higher migratory activity than treatment at 37°C. Invasive capacity was increased after heat treatment, ranging from 115% to 126%. Expression of several epithelial-mesenchymal transition markers, including ZEB1, N-cadherin, and MMP2, was upregulated following heat treatment at 40°C. CONCLUSION We for the first time demonstrate that sublethal thermal stress may increase clonogenicity, migration, and invasion of thyroid cancer cells.
Collapse
Affiliation(s)
- Chi-Yu Kuo
- Department of Surgery, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, School of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Chung-Hsin Tsai
- Department of Surgery, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, School of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Jun Kui Wu
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Shih-Ping Cheng
- Department of Surgery, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, School of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan
| |
Collapse
|
5
|
Ye F, Xie L, Liang L, Zhou Z, He S, Li R, Lin L, Zhu K. Mechanisms and therapeutic strategies to combat the recurrence and progression of hepatocellular carcinoma after thermal ablation. J Interv Med 2023; 6:160-169. [PMID: 38312128 PMCID: PMC10831380 DOI: 10.1016/j.jimed.2023.10.004] [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: 08/20/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 02/06/2024] Open
Abstract
Thermal ablation (TA), including radiofrequency ablation (RFA) and microwave ablation (MWA), has become the main treatment for early-stage hepatocellular carcinoma (HCC) due to advantages such as safety and minimal invasiveness. However, HCC is prone to local recurrence, with more aggressive malignancies after TA closely related to TA-induced changes in epithelial-mesenchymal transition (EMT) and remodeling of the tumor microenvironment (TME). According to many studies, various components of the TME undergo complex changes after TA, such as the recruitment of innate and adaptive immune cells, the release of tumor-associated antigens (TAAs) and various cytokines, the formation of a hypoxic microenvironment, and tumor angiogenesis. Changes in the TME after TA can partly enhance the anti-tumor immune response; however, this response is weak to kill the tumor completely. Certain components of the TME can induce an immunosuppressive microenvironment through complex interactions, leading to tumor recurrence and progression. How the TME is remodeled after TA and the mechanism by which the TME promotes HCC recurrence and progression are unclear. Thus, in this review, we focused on these issues to highlight potentially effective strategies for reducing and preventing the recurrence and progression of HCC after TA.
Collapse
Affiliation(s)
| | | | | | - Zhimei Zhou
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Siqin He
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Rui Li
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Liteng Lin
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Kangshun Zhu
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| |
Collapse
|
6
|
Wang F, Xu C, Li G, Lv P, Gu J. Incomplete radiofrequency ablation induced chemoresistance by up-regulating heat shock protein 70 in hepatocellular carcinoma. Exp Cell Res 2021; 409:112910. [PMID: 34801560 DOI: 10.1016/j.yexcr.2021.112910] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/18/2021] [Accepted: 11/05/2021] [Indexed: 12/19/2022]
Abstract
Radiofrequency ablation (RFA) is a common minimally invasive treatment for hepatocellular carcinoma (HCC). Incomplete RFA (iRFA) due to the sub-lethal heat shock challenge of some cell populations leads to the generation of transformed survivor cells with enhanced chemoresistance. However, the underlying mechanism of iRFA on HCCs chemoresistance remains unknown. In the present study, we investigated the effect of iRFA on HCCs sensitivity to cisplatin. Cells treated with the sub-lethal heat shock challenge were used to mimic iRFA treatment in vitro. An orthotopic implantation HCC model was established and also performed iRFA treatment. Flow cytometry, transwell assay, and cell counting kit-8 assay were used to determine the effect of iRFA treatment on cisplatin-induced HCC cell apoptosis, invasion, and cell viability. ELISA and Western blot were used to detect the effect of iRFA treatment on cisplatin-induced HCC cell pyroptosis. We found that iRFA treatment increased the HCC cell proliferation and invasion ability, and inhibited cisplatin-induced pyroptosis. Further experiments showed that iRFA treatment induced upregulation of HSP70, which inhibited the cisplatin-induced NLRP3 inflammasome activation, leading to inhibition of pyroptosis. HSP70 knockdown or NLRP3 overexpression could reverse the effect of iRFA treatment in vitro. In vivo, HSP70 knockdown reversed the chemosensitivity of HCC to cisplatin, which was decreased by iRFA. In conclusion, we demonstrated that iRFA induced drug resistance by HSP70-mediated inhibition of cell pyroptosis in HCC.
Collapse
Affiliation(s)
- Fuan Wang
- Department of Interventional Radiology, Northern Jiangsu People's Hospital (Clinical Medical College, Yangzhou University), 98 Nantong West Road, Yangzhou, JiangSu Province, China; Vascular and Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, PR China
| | - Chuan Xu
- Radiology, Clinical Medical of Shanghai Tenth People's Hospital of Nanjing Medical University, Yanchang Road 301#, Shanghai, China
| | - Guiling Li
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital (Clinical Medical College, Yangzhou University), 98 Nantong West Road, Yangzhou, JiangSu Province, China.
| | - Penghua Lv
- Department of Interventional Radiology, Northern Jiangsu People's Hospital (Clinical Medical College, Yangzhou University), 98 Nantong West Road, Yangzhou, JiangSu Province, China
| | - Jianping Gu
- Vascular and Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, PR China.
| |
Collapse
|
7
|
Zhang YJ, Chen MS, Chen Y, Lau WY, Peng Z. Long-term Outcomes of Transcatheter Arterial Chemoembolization Combined With Radiofrequency Ablation as an Initial Treatment for Early-Stage Hepatocellular Carcinoma. JAMA Netw Open 2021; 4:e2126992. [PMID: 34570206 PMCID: PMC8477266 DOI: 10.1001/jamanetworkopen.2021.26992] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IMPORTANCE The long-term outcomes of transcatheter arterial chemoembolization (TACE) combined with radiofrequency ablation (RFA) are not determined. OBJECTIVE To report the long-term outcomes of TACE-RFA. DESIGN, SETTING, AND PARTICIPANTS This cohort study analyzed long-term follow-up data from a phase 3 randomized clinical trial of adults with early HCC conducted from October 2006 to June 2009. Participants were randomly assigned to the TACE-RFA group or the RFA group in a 1:1 ratio and followed up approximately 6 years after the trial was closed. Data analysis was performed March 2020. EXPOSURE In the TACE-RFA group, TACE was performed first, and RFA was done 2 weeks later. MAIN OUTCOMES AND MEASURES Overall survival (OS) and recurrence-free survival (RFS). RESULTS Of 189 patients who were included (mean [SD] age, 54.3 [12.0] years; 146 [77.2%] men), 94 and 95 patients were assigned to the TACE-RFA group and RFA group, respectively, with their baseline characteristics well matched. Three patients in each group were lost to follow-up. The 5-year and 7-year OS rates for the TACE-RFA group vs the RFA group were 52.0% and 36.4% vs 43.2% and 19.4%, respectively (hazard ratio [HR], 0.55; 95% CI, 0.39-0.78; P = .001). The 5-year and 7-year RFS rates for the TACE-RFA group vs the RFA group were 41.4% and 34.5% vs 27.4% and 18.1%, respectively (HR, 0.66; 95% CI, 0.49-0.89; P = .007). On subgroup analysis comparing patients who had tumors larger than 3 cm with those who had tumors 3 cm or smaller, the OS and RFS survival rates in the TACE-RFA group (HR, 3.20; 95% CI, 1.91-5.35, P < .001) were significantly better than those in the RFA group (HR, 2.03; 95% CI, 1.30-3.17; P = .002). CONCLUSIONS AND RELEVANCE In this cohort study, combined RFA and TACE was associated with better survival than RFA alone on long-term follow-up. Patients with tumors 3 cm or smaller did not benefit as well as patients with tumors larger than 3 cm from the combined treatment.
Collapse
Affiliation(s)
- Yao Jun Zhang
- Department of Hepatobiliary Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
| | - Min Shan Chen
- Department of Hepatobiliary Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
| | - Yong Chen
- Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wan Yee Lau
- Faculty of Medicine, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR, China
| | - Zhenwei Peng
- Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Su T, Huang M, Liao J, Lin S, Yu P, Yang J, Cai Y, Zhu S, Xu L, Peng Z, Peng S, Chen S, Kuang M. Insufficient Radiofrequency Ablation Promotes Hepatocellular Carcinoma Metastasis Through N6-Methyladenosine mRNA Methylation-Dependent Mechanism. Hepatology 2021; 74:1339-1356. [PMID: 33638162 DOI: 10.1002/hep.31766] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 01/06/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS The dynamic N6-methyladenosine (m6 A) mRNA modification is essential for acute stress response and cancer progression. Sublethal heat stress from insufficient radiofrequency ablation (IRFA) has been confirmed to promote HCC progression; however, whether m6 A machinery is involved in IRFA-induced HCC recurrence remains open for study. APPROACH AND RESULTS Using an IRFA HCC orthotopic mouse model, we detected a higher level of m6 A reader YTH N6-methyladenosine RNA binding protein 1-3 (YTHDF1) in the sublethal-heat-exposed transitional zone close to the ablation center than that in the farther area. In addition, we validated the increased m6 A modification and elevated YTHDF1 protein level in sublethal-heat-treated HCC cell lines, HCC patient-derived xenograft (PDX) mouse model, and patients' HCC tissues. Functionally, gain-of-function/loss-of-function assays showed that YTHDF1 promotes HCC cell viability and metastasis. Knockdown of YTHDF1 drastically restrains the tumor metastasis evoked by sublethal heat treatment in tail vein injection lung metastasis and orthotopic HCC mouse models. Mechanistically, we found that sublethal heat treatment increases epidermal factor growth receptor (EGFR) m6 A modification in the vicinity of the 5' untranslated region and promotes its binding with YTHDF1, which enhances the translation of EGFR mRNA. The sublethal-heat-induced up-regulation of EGFR level was further confirmed in the IRFA HCC PDX mouse model and patients' tissues. Combination of YTHDF1 silencing and EGFR inhibition suppressed the malignancies of HCC cells synergically. CONCLUSIONS The m6 A-YTHDF1-EGFR axis promotes HCC progression after IRFA, supporting the rationale for targeting m6 A machinery combined with EGFR inhibitors to suppress HCC metastasis after RFA.
Collapse
Affiliation(s)
- Tianhong Su
- Department of Liver Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Manling Huang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junbin Liao
- Department of Liver Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuibin Lin
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peng Yu
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianhua Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuhong Cai
- Department of Liver Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shenghua Zhu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lixia Xu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhenwei Peng
- Clinical Trials Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sui Peng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Clinical Trials Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuling Chen
- Division of Interventional Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming Kuang
- Department of Liver Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
ATP-citrate lyase regulates stemness and metastasis in hepatocellular carcinoma via the Wnt/β-catenin signaling pathway. Hepatobiliary Pancreat Dis Int 2021; 20:251-261. [PMID: 33129711 DOI: 10.1016/j.hbpd.2020.05.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/29/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most highly malignant tumors. Liver tumor-initiating cells (LTICs) have been considered to contribute to HCC progression and metastasis. ATP-citrate lyase (ACLY), as a key enzyme for de novo lipogenesis, has been reported to be upregulated in various tumors. However, its expression and role in HCC and LTICs remain unknown. METHODS The expressions of ACLY in HCC tissues were detected by quantitative real-time PCR (qRT-PCR), Western blotting and immunohistochemistry. Kaplan-Meier curves and Chi-square test were used to determine the clinical significance of ACLY expression in HCC patients. A series of assays were performed to determine the function of ACLY on stemness, migration and invasion of HCC cells. Luciferase reporter assay, Western blotting and immunoprecipitation were used to study the regulation of the Wnt/β-catenin signaling by ACLY. Rescue experiments were performed to investigate whether β-catenin was the mediator of ACLY-regulated stemness and migration in HCC cells. RESULTS ACLY was highly expressed in HCC tissues and LTICs. Overexpression of ACLY was significantly correlated with poor prognosis, progression and metastasis of HCC patients. Knockdown of ACLY remarkably suppressed stemness properties, migration and invasion in HCC cells. Mechanistically, ACLY could regulate the canonical Wnt pathway by affecting the stability of β-catenin, and Lys49 acetylation of β-catenin might mediate ACLY-regulated β-catenin level in HCC cells. CONCLUSIONS ACLY is a potent regulator of Wnt/β-catenin signaling in modulating LTICs stemness and metastasis in HCC. ACLY may serve as a new target for the diagnosis and treatment of HCC.
Collapse
|
10
|
Zhou G, Da Won Bae S, Nguyen R, Huo X, Han S, Zhang Z, Hebbard L, Duan W, Eslam M, Liddle C, Yuen L, Lam V, Qiao L, George J. An aptamer-based drug delivery agent (CD133-apt-Dox) selectively and effectively kills liver cancer stem-like cells. Cancer Lett 2020; 501:124-132. [PMID: 33352247 DOI: 10.1016/j.canlet.2020.12.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/02/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
Liver cancer has no effective therapies, hence a poor survival. Cancer stem-like cells not only contribute to cancer initiation and progression, but also to drug resistance, cancer metastasis, and eventually treatment failure. Hence, any approaches that can effectively kill cancer stem-like cells hold a great potential for cancer treatment. CD133 is a robust marker for liver cancer stem-like cells. We developed a specific aptamer against CD133 (CD133-apt), and then loaded this aptamer with an anticancer drug doxorubicin (CD133-apt-Dox). The efficacy of CD133-apt-Dox in targeting liver cancer stem-like cells and its overall effect in treating liver cancer were investigated using multiple in vitro and in vivo studies including in patients-derived liver cancer organoids. We have observed that CD133-apt could preferably delivered doxorubicin to CD133-expressing cells with efficient drug accumulation and retention. CD133-apt-Dox impaired the self-renewal capacity of liver cancer stem-like cells and attenuated their stem-ness phenotypes in vitro or in vivo. CD133-apt-Dox significantly inhibited the growth of liver cancer cells and patients-derived organoids and reduced the growth of xenograft tumours in nude mice inhibited the growth of DEN-induced liver cancer in immunocompetent mice. Hence, aptamer-mediated targeting of CD133 is a highly promising approach for liver cancer therapy.
Collapse
MESH Headings
- AC133 Antigen/genetics
- Animals
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/pharmacokinetics
- Aptamers, Nucleotide/administration & dosage
- Aptamers, Nucleotide/genetics
- Aptamers, Nucleotide/pharmacokinetics
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Doxorubicin/administration & dosage
- Doxorubicin/pharmacokinetics
- Drug Carriers/administration & dosage
- Drug Carriers/pharmacokinetics
- Drug Delivery Systems/methods
- HEK293 Cells
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
Collapse
Affiliation(s)
- Gang Zhou
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Sarah Da Won Bae
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Romario Nguyen
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Xiaoqi Huo
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Shuanglin Han
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Zhiqiang Zhang
- Renal Inflammation and Immunology Group, Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia; Department of Urology, The Second Affiliated Hospital of Anhui Medical University, 230601, Anhui, China
| | - Lionel Hebbard
- Discipline of Molecular and Cell Biology, Australian Institute for Tropical Health and Medicine, Centre for Molecular Therapeutics, James Cook University, Townsville, 4811, Australia
| | - Wei Duan
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria, 3217, Australia
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Lawrence Yuen
- Department of Surgery, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Vincent Lam
- Department of Surgery, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia.
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia.
| |
Collapse
|
11
|
c-Met/MAPK pathway promotes the malignant progression of residual hepatocellular carcinoma cells after insufficient radiofrequency ablation. Med Oncol 2020; 37:117. [PMID: 33215351 DOI: 10.1007/s12032-020-01444-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 11/14/2020] [Indexed: 12/17/2022]
Abstract
Radiofrequency ablation (RFA) is popularly used in the treatment of hepatocellular carcinoma (HCC). However, the accelerated malignant progression of residual HCC cells after RFA is the main obstacle for the application of this technology in HCC treatment. In the present study, HepG2 cells, an established human HCC cell line, experienced repeatedly with heat treatment, survived cells, HepG2-H cells, were used to simulate residual HCC cells after RFA. The abilities of proliferation, colony formation, and migration were compared between HepG2 and HepG2-H cells. Then, RNA sequencing was used to explore the difference in genes expression between two groups of cells. Subsequently, the level of c-Met, one of membranous receptors of MAPK signal pathway, was measured by RT-qPCR and western blot; the effect of c-Met inhibition on the malignant progression of HepG2-H cells was evaluated. The results showed that HepG2-H cells exhibited higher abilities in the proliferation, colony formation, and migration than that of HepG2 cells. Moreover, differentially expressed genes between two groups of cells were prominently enriched in MAPK signal pathway. The level of c-Met in HepG2-H cells was significantly higher than that in HepG2 cells, and the inhibition in the activity of c-Met could repress the malignant behaviors of HepG2-H cells. These results indicated that the accelerated malignant progression of residual HCC cells after RFA can be partly attributed to the overexpression of c-Met and the activation of MAPK signal pathway. Therefore, we proposed that RFA followed by c-Met inhibitor intake maybe is a better treatment protocol for HCC.
Collapse
|
12
|
Zhao Y, Li K, Sun J, He N, Zhao P, Zang C, Yang X, Hu C, Long J, Zhang H, Wang Q, Zhao Y, Zhang Y. Genomic DNA methylation profiling indicates immune response following thermal ablation treatment for HBV-associated hepatocellular carcinoma. Oncol Lett 2020; 20:677-684. [PMID: 32565992 PMCID: PMC7285841 DOI: 10.3892/ol.2020.11636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 04/15/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatitis B virus (HBV)-associated hepatocellular carcinoma (HCC) is the most common type of liver cancer in China. Thermal ablation is one of the main strategies for HCC treatment. However, few studies have investigated the properties of the immune response following thermal ablation thus far. In the present study, five subjects with HBV-associated HCC were recruited from The Beijing Youan Hospital. Peripheral blood mononuclear cells (PBMCs) were collected at three time points: Prior to thermal ablation (PR), 1-3 days post-ablation (P1) and 5-7 days post-ablation (P7). An Illumina 850K methylation microarray was employed to determine the DNA methylation profile of each sample. Data were analyzed using different methylation probes with the Bioconductor package in R. Following annotation of different methylation CG sites (CGs), the associated genes were subjected to an Ingenuity Pathway Analysis. A total of 3,000 significantly different CGs (adjusted P<0.05; |log(fold-change)|>0.5) were identified within the PR, P1 and P7 time points. Of these, 744 (24.8%) sites increased between the PR and P1 time points but gradually decreased at the P7 time point. The remaining 2,256 (75.2%) sites decreased between the PR and P1 time points gradually increased at the P7 time point. Following gene annotation of different CGs on the promoter, signaling pathways analysis demonstrated that 'p70S6K signaling', 'CXCR4 signaling', 'dendritic cell maturation', 'production of nitric oxide and reactive oxygen species in macrophages' pathways were activated at the P7 time point. The present study suggested that PBMC DNA methylation had changed soon after thermal ablation for subjects with HBV-associated HCC, and systemic immune responses were activated, particularly at the P7 time point.
Collapse
Affiliation(s)
- Yanan Zhao
- Research Center for Biomedical Resources, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China.,Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Kang Li
- Research Center for Biomedical Resources, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Jianping Sun
- Research Center for Biomedical Resources, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Ning He
- Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Peng Zhao
- Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Chaoran Zang
- Research Center for Biomedical Resources, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China.,Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Xiaozhen Yang
- Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Caixia Hu
- Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Jiang Long
- Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Honghai Zhang
- Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Qi Wang
- Research Center for Biomedical Resources, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China.,Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Yan Zhao
- Clinical Detection Center, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Yonghong Zhang
- Research Center for Biomedical Resources, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China.,Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
13
|
Partridge B, Rossmeisl JH, Kaloss AM, Basso EKG, Theus MH. Novel ablation methods for treatment of gliomas. J Neurosci Methods 2020; 336:108630. [PMID: 32068011 DOI: 10.1016/j.jneumeth.2020.108630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 12/18/2022]
Abstract
Primary brain tumors are among the deadliest cancers that remain highly incurable. A need exists for new approaches to tumor therapy that can circumvent the blood brain barrier (BBB), target highly resistant tumors and cancer stem-like cells (CSCs) as well create an anti-cancer immunomodulatory environment. Successful treatments may also require a combinatory approach utilizing surgery, chemotherapy, radiation and novel ablation strategies that can both eliminate the bulk tumor and prevent any potential residual CSCs from propagating in the resected tissue. A number of thermal and non-thermal ablation methods have been developed and tested, which have gained much enthusiasm for the treatment of brain tumors. Here we review the most common primary brain tumors and the candidate ablation methods for targeting the tumor and its microenvironment.
Collapse
Affiliation(s)
- Brittanie Partridge
- Veterinary and Comparative Neuro-oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - John H Rossmeisl
- Veterinary and Comparative Neuro-oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Alexandra M Kaloss
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Erwin Kristobal Gudenschwager Basso
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; School of Neuroscience, Virginia Tech, Blacksburg VA 24061, USA; Center for Regenerative Medicine, VT College of Veterinary Medicine, Blacksburg, Virginia, 24061, USA.
| |
Collapse
|