1
|
Zhu L, Ren Y, Dong M, Sun B, Huang J, Chen L, Xia X, Dong X, Zheng C. Ultrasmall Metal TPZ Complexes with Deep Tumor Penetration for Enhancing Radiofrequency Ablation Therapy and Inducing Antitumor Immune Responses. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311244. [PMID: 38898764 DOI: 10.1002/smll.202311244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/10/2024] [Indexed: 06/21/2024]
Abstract
Radiofrequency ablation (RFA) is one of the most common minimally invasive techniques for the treatment of solid tumors, but residual malignant tissues or small satellite lesions after insufficient RFA (iRFA) are difficult to remove, often leading to metastasis and recurrence. Here, Fe-TPZ nanoparticles are designed by metal ion and (TPZ) ligand complexation for synergistic enhancement of RFA residual tumor therapy. Fe-TPZ nanoparticles are cleaved in the acidic microenvironment of the tumor to generate Fe2+ and TPZ. TPZ, an anoxia-dependent drug, is activated in residual tumors and generates free radicals to cause tumor cell death. Elevated Fe2+ undergoes a redox reaction with glutathione (GSH), inducing a strong Fenton effect and promoting the production of the highly toxic hydroxyl radical (•OH). In addition, the ROS/GSH imbalance induced by this treatment promotes immunogenic cell death (ICD), which triggers the release of damage-associated molecular patterns, macrophage polarization, and lymphocyte infiltration, thus triggering a systemic antitumor immune response and noteworthy prevention of tumor metastasis. Overall, this integrated treatment program driven by multiple microenvironment-dependent pathways overcomes the limitations of the RFA monotherapy approach and thus improves tumor prognosis. Furthermore, these findings aim to provide new research ideas for regulating the tumor immune microenvironment.
Collapse
Affiliation(s)
- Licheng Zhu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yanqiao Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Mengna Dong
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bo Sun
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jia Huang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lei Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiangwen Xia
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiangjun Dong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| |
Collapse
|
2
|
Guo J, Yan Y, Sun J, Ji K, Hei Z, Zeng L, Xu H, Ren X, Sun Y. Chaperones Hsc70 and Hsp70 play distinct roles in the replication of bocaparvovirus minute virus of canines. Mol Microbiol 2024; 121:1127-1147. [PMID: 38629786 DOI: 10.1111/mmi.15263] [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: 09/27/2023] [Revised: 03/09/2024] [Accepted: 04/02/2024] [Indexed: 06/14/2024]
Abstract
Minute virus of canines (MVC) belongs to the genus Bocaparvovirus (formerly Bocavirus) within the Parvoviridae family and causes serious respiratory and gastrointestinal symptoms in neonatal canines worldwide. A productive viral infection relies on the successful recruitment of host factors for various stages of the viral life cycle. However, little is known about the MVC-host cell interactions. In this study, we identified that two cellular proteins (Hsc70 and Hsp70) interacted with NS1 and VP2 proteins of MVC, and both two domains of Hsc70/Hsp70 were mediated for their interactions. Functional studies revealed that Hsp70 was induced by MVC infection, knockdown of Hsc70 considerably suppressed MVC replication, whereas the replication was dramatically promoted by Hsp70 knockdown. It is interesting that low amounts of overexpressed Hsp70 enhanced viral protein expression and virus production, but high amounts of Hsp70 overexpression weakened them. Upon Hsp70 overexpressing, we observed that the ubiquitination of viral proteins changed with Hsp70 overexpression, and proteasome inhibitor (MG132) restored an accumulation of viral proteins. In addition, we verified that Hsp70 family inhibitors remarkably decreased MVC replication. Overall, we identified Hsc70 and Hsp70 as interactors of MVC NS1 and VP2 proteins and were involved in MVC replication, which may provide novel targets for anti-MVC approach.
Collapse
Affiliation(s)
- Jianhui Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Yan Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Jinhan Sun
- Department of Clinical Medicine, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Kai Ji
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Zhiping Hei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Liang Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Huanzhou Xu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiang Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Yuning Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
3
|
Sojka DR, Gogler A, Kania D, Vydra N, Wiecha K, Adamiec-Organiściok M, Wilk A, Chumak V, Matyśniak D, Scieglinska D. The human testis-enriched HSPA2 interacts with HIF-1α in epidermal keratinocytes, yet HIF-1α stability and HIF-1-dependent gene expression rely on the HSPA (HSP70) activity. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119735. [PMID: 38641179 DOI: 10.1016/j.bbamcr.2024.119735] [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: 08/17/2023] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 04/21/2024]
Abstract
The Hypoxia-Inducible Factor 1 (HIF-1) is essential for cellular adaptation to reduced oxygen levels. It also facilitates the maintenance and re-establishment of skin homeostasis. Among others, it is involved in regulating keratinocyte differentiation. The stability of the oxygen-liable HIF-1α subunit is regulated by various non-canonical oxygen-independent mechanisms, which among others involve Heat Shock Proteins of the A family (HSPA/HSP70). This group of highly homologous chaperones and proteostasis-controlling factors includes HSPA2, a unique member crucial for spermatogenesis and implicated in the regulation of keratinocyte differentiation. HIF-1 can control the HSPA2 gene expression. In this study, we revealed that HIF-1α is the first confirmed client of HSPA2 in human somatic cells. It colocalises and interacts directly with HSPA2 in the epidermis in situ and immortalised keratinocytes in vitro. Using an in vitro model based on HSPA2-overexpressing and HSPA2-deficient variants of immortalised keratinocytes we showed that changes in HSPA2 levels do not affect the levels and intracellular localisation of HIF-1α or influence the ability of HIF-1 to modulate target gene expression. However, HIF-1α stability in keratinocytes appears critically reliant on HSPAs as a group of functionally overlapping chaperones. In addition to HSPA2, HIF-1α colocalises and forms complexes with HSPA8 and HSPA1, representing housekeeping and stress-inducible HSPA family paralogs, respectively. Chemical inhibition of HSPA activity, but not paralog-specific knockdown of HSPA8 or HSPA1 expression reduced HIF-1α levels and HIF-1-dependent gene expression. These observations suggest that pharmacological targeting of HSPAs could prevent excessive HIF-1 signalling in pathological skin conditions.
Collapse
Affiliation(s)
- Damian Robert Sojka
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland
| | - Agnieszka Gogler
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland
| | - Daria Kania
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland
| | - Natalia Vydra
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland
| | - Klaudia Wiecha
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland
| | - Małgorzata Adamiec-Organiściok
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland; Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland
| | - Agata Wilk
- Department of Biostatistics and Bioinformatics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland; Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland
| | - Vira Chumak
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland
| | - Damian Matyśniak
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland
| | - Dorota Scieglinska
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland.
| |
Collapse
|
4
|
Ao F, Li X, Tan Y, Jiang Z, Yang F, Guo J, Zhu Q, Chen Z, Zhou B, Zhang K, Li D. STING agonist-based hydrogel enhances immune activation in synergy with radiofrequency ablation for hepatocellular carcinoma treatment. J Control Release 2024; 369:296-308. [PMID: 38301925 DOI: 10.1016/j.jconrel.2024.01.048] [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: 08/03/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024]
Abstract
Immunosuppression caused by incomplete radiofrequency ablation (iRFA) is a crucial factor affecting the effectiveness of RFA for solid tumors. However, little is known about the changes iRFA induces in the tumor immune microenvironment (TIME) of hepatocellular carcinoma (HCC), the primary application area for RFA. In this study, we found iRFA promotes a suppressive TIME in residual HCC tumors, characterized by M2 macrophage polarization, inhibited antigen presentation by dendritic cells (DCs), and reduced infiltration of cytotoxic T lymphocytes (CTLs). Interestingly, the STING agonist MSA-2 was able to reorganize M2-like tumor-promoting macrophages into M1-like anti-tumor states and enhance antigen presentation by DCs. To optimize the therapeutic effect of MSA-2, we used a calcium ion (Ca2+) responsive sodium alginate (ALG) as a carrier, forming an injectable hydrogel named ALG@MSA-2. This hydrogel can change from liquid to gel, maintaining continuous drug release in situ. Our results suggested that ALG@MSA-2 effectively activated anti-tumor immunity, as manifested by increased M1-like macrophage polarization, enhanced antigen presentation by DCs, increased CTL infiltration, and inhibited residual tumor growth. ALG@MSA-2 also resulted in a complete regression of contralateral tumors and widespread liver metastases in vivo. In addition, the excellent biosafety of ALG@MSA-2 was also proved by blood biochemical analysis and body weight changes in mice. In summary, this study demonstrated that the immune cascade of ALG@MSA-2 mediated the STING pathway activation and promoted a favorable TIME which might provide novel insights for the RFA treatment of HCC.
Collapse
Affiliation(s)
- Feng Ao
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Nuclear Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Xi Li
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Yan Tan
- Department of Nuclear Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zebo Jiang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Fan Yang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Pediatrics, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, Guangdong Province, China
| | - Jingpei Guo
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Qiancheng Zhu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zhongguo Chen
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Bin Zhou
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China.
| | - Ke Zhang
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China.
| | - Dan Li
- Department of Nuclear Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China.
| |
Collapse
|
5
|
Shi X, Liu C, Zheng W, Cao X, Li W, Zhang D, Zhu J, Zhang X, Chen Y. Proteomic Analysis Revealed the Potential Role of MAGE-D2 in the Therapeutic Targeting of Triple-Negative Breast Cancer. Mol Cell Proteomics 2024; 23:100703. [PMID: 38128647 PMCID: PMC10835320 DOI: 10.1016/j.mcpro.2023.100703] [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: 03/05/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Among all the molecular subtypes of breast cancer, triple-negative breast cancer (TNBC) is the most aggressive one. Currently, the clinical prognosis of TNBC is poor because there is still no effective therapeutic target. Here, we carried out a combined proteomic analysis involving bioinformatic analysis of the proteome database, label-free quantitative proteomics, and immunoprecipitation (IP) coupled with mass spectrometry (MS) to explore potential therapeutic targets for TNBC. The results of bioinformatic analysis showed an overexpression of MAGE-D2 (melanoma antigen family D2) in TNBC. In vivo and in vitro experiments revealed that MAGE-D2 overexpression could promote cell proliferation and metastasis. Furthermore, label-free quantitative proteomics revealed that MAGE-D2 acted as a cancer-promoting factor by activating the PI3K-AKT pathway. Moreover, the outcomes of IP-MS and cross-linking IP-MS demonstrated that MAGE-D2 could interact with Hsp70 and prevent Hsp70 degradation, but evidence for their direct interaction is still lacking. Nevertheless, MAGE-D2 is a potential therapeutic target for TNBC, and blocking MAGE-D2 may have important therapeutic implications.
Collapse
Affiliation(s)
- Xiaoyu Shi
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Chunyan Liu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Weimin Zheng
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xiao Cao
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Wan Li
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Dongxue Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Jianhua Zhu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xian Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing, China; Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Nanjing, China.
| |
Collapse
|
6
|
Peng B, Ling X, Huang T, Wan J. HSP70 via HIF-1 α SUMOylation inhibits ferroptosis inducing lung cancer recurrence after insufficient radiofrequency ablation. PLoS One 2023; 18:e0294263. [PMID: 37948404 PMCID: PMC10637661 DOI: 10.1371/journal.pone.0294263] [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: 03/03/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Radiofrequency ablation (RFA) is an effective and feasible therapy for lung cancer, but accelerated progression of residual non-small cell lung cancer (NSCLC) after incomplete radiofrequency ablation (RFA) has frequently been reported. A previous study reported that HSP70 and HIF-1α were highly expressed in areas with incomplete RFA. Therefore, we sought to elucidate the regulatory effect of the HIF-1α/HSP70 pathway on lung cancer recurrence after incomplete radiofrequency ablation. In this study, we found that knockdown of HSP70 can reduce sumo 1, sumo 2/3 (marker of SUMOylation) of HIF-1α and inhibit A549 cell proliferation and migration under heat stress conditions (used to simulate incomplete RFA in vitro). We observed that knockdown of HSP70 altered the expression of ferroptosis-related proteins and genes (SLC7A11 and ACSL3), and the RNA-seq results showed that knockdown of HSP70 activated the ferroptosis pathway, further confirming that HSP70 regulates ferroptosis. In summary, HSP70, via HIF-1α SUMOylation, inhibited ferroptosis, inducing lung cancer recurrence after radiofrequency ablation. The study reveals a new direction for further research on therapeutic targets to suppress lung cancer recurrence and provides a theoretical foundation for further clinical studies.
Collapse
Affiliation(s)
- Bin Peng
- Department of Thoracic Surgery, Shenzhen People’s Hospital, Shenzhen, China
| | - Xiean Ling
- Department of Thoracic Surgery, Shenzhen People’s Hospital, Shenzhen, China
| | - Tonghai Huang
- Department of Thoracic Surgery, Shenzhen People’s Hospital, Shenzhen, China
| | - Jun Wan
- Department of Thoracic Surgery, Shenzhen People’s Hospital, Shenzhen, China
| |
Collapse
|
7
|
Chen C, Han Q, Ren H, Wu S, Li Y, Guo J, Li X, Liu X, Li C, Tian Y. Multiparametric MRI-based model for prediction of local progression of hepatocellular carcinoma after thermal ablation. Cancer Med 2023; 12:17529-17540. [PMID: 37694337 PMCID: PMC10524055 DOI: 10.1002/cam4.6277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/06/2023] [Accepted: 06/11/2023] [Indexed: 09/12/2023] Open
Abstract
PURPOSE To develop a deep learning radiomics of multiparametric magnetic resonance imaging (DLRMM)-based model that incorporates preoperative and postoperative signatures for prediction of local tumor progression (LTP) after thermal ablation (TA) in hepatocellular carcinoma (HCC). METHODS From May 2017 to October 2021, 417 eligible patients with HCC were retrospectively enrolled from three hospitals (one primary cohort [PC, n = 189] and two external test cohorts [ETCs][n = 135, 93]). DLRMM features were extracted from T1WI + C, T2WI, and DWI using ResNet18 model. An integrative model incorporating the DLRMM signature with clinicopathologic variables were further built to LTP risk stratification. The performance of these models were compared by areas under receiver operating characteristic curve (AUC) using DeLong test. RESULTS A total of 1668 subsequences and 31,536 multiparametric MRI slice including T1WI, T2WI, and DWI were collected simultaneously. The DLRMM signatures were extracted from tumor and ablation zone, respectively. Ablative margin, multiple tumors, and tumor abutting major vessels were regarded as risk factors for LTP in clinical model. The AUC of DLRMM model were 0.864 in PC, 0.843 in ETC1, and 0.858 in ETC2, which was higher significantly than those in clinical model (p < 0.001). After integrating clinical variable, DLRMM model obtained significant improvement with AUC of 0.870-0.869 in three cohorts (all, p < 0.001), which can provide the risk stratification for overall survival of HCC patients. CONCLUSIONS The DLRMM model is essential to identify LTP risk of HCC patients who underwent TA and may potentially benefit personalized decision-making.
Collapse
Affiliation(s)
- Chao Chen
- Department of Minimal Invasive Intervention RadiologyGanzhou People's HospitalGanzhouChina
| | - Qiuying Han
- Department of CardiologyThe First Affiliated Hospital of Jinan universityGuanghzhouChina
| | - He Ren
- Department of UltrasoundThe Six Medical Center of Chinese PLA General HospitalBeijingChina
| | - Siyi Wu
- Department of Interventional Radiology and Vascular SurgeryThe First Affiliated Hospital of Jinan UniversityGuanghzhouChina
| | - Yangyang Li
- Department of Interventional Radiology and Vascular SurgeryThe First Affiliated Hospital of Jinan UniversityGuanghzhouChina
| | - Jiandong Guo
- Department of Interventional Radiology and Vascular SurgeryThe First Affiliated Hospital of Jinan UniversityGuanghzhouChina
| | - Xinghai Li
- Department of Minimal Invasive Intervention RadiologyGanzhou People's HospitalGanzhouChina
| | - Xiang Liu
- Department of Minimal Invasive Intervention RadiologyGanzhou People's HospitalGanzhouChina
| | - Chengzhi Li
- Department of Interventional Radiology and Vascular SurgeryThe First Affiliated Hospital of Jinan UniversityGuanghzhouChina
| | - Yunfei Tian
- Department of Minimal Invasive Intervention RadiologyGanzhou People's HospitalGanzhouChina
| |
Collapse
|
8
|
How Biology Guides the Combination of Locoregional Interventional Therapies and Immunotherapy for Hepatocellular Carcinoma: Cytokines and Their Roles. Cancers (Basel) 2023; 15:cancers15041324. [PMID: 36831664 PMCID: PMC9954096 DOI: 10.3390/cancers15041324] [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: 01/10/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
As most patients with hepatocellular carcinoma (HCC) are diagnosed at the intermediate or advanced stage and are no longer eligible for curative treatment, the overall survival rate of HCC remains unsatisfactory. Locoregional interventional therapies (LITs), and immune checkpoint inhibitor (ICI)-based immunotherapy, focus on treating HCC, but the efficacy of their individual application is limited. Therefore, the purpose of this review was to discuss the biological roles of cytokines and their therapeutic potential in the combination therapy of LITs and ICI-based immunotherapy. The two common techniques of LITs are ablative and transarterial therapies. Whether LITs are complete or incomplete can largely affect the antitumor immune response and tumor progression. Cytokines that induce both local and systemic responses to LITs, including interferons, interleukins, chemokines, TNF-α, TGF-β, VEGF, and HGF, and their roles are discussed in detail. In addition, specific cytokines that can be used as therapeutic targets to reduce immune-related adverse events (irAEs) are introduced. Overall, incomplete LITs in a tumor, combined with specific cytokines, are thought to be effective at improving the therapeutic efficacy and reducing treatment-induced irAEs, and represent a new hope for managing unresectable HCC.
Collapse
|
9
|
Xiao C, Liu S, Ge G, Jiang H, Wang L, Chen Q, Jin C, Mo J, Li J, Wang K, Zhang Q, Zhou J. Roles of hypoxia-inducible factor in hepatocellular carcinoma under local ablation therapies. Front Pharmacol 2023; 14:1086813. [PMID: 36814489 PMCID: PMC9939531 DOI: 10.3389/fphar.2023.1086813] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/18/2023] [Indexed: 02/09/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common digestive malignancies. HCC It ranges as the fifth most common cause of cancer mortality worldwide. While The prognosis of metastatic or advanced HCC is still quite poor. Recently, locoregional treatment, especially local ablation therapies, plays an important role in the treatment of HCC. Radiofrequency ablation (RFA) and high-intensity focused ultrasound (HIFU) ablation are the most common-used methods effective and feasible for treating HCC. However, the molecular mechanisms underlying the actions of ablation in the treatments for HCC and the HCC recurrence after ablation still are poorly understood. Hypoxia-inducible factor (HIF), the key gene switch for adaptive responses to hypoxia, has been found to play an essential role in the rapid aggressive recurrence of HCC after ablation treatment. In this review, we summarized the current evidence of the roles of HIF in the treatment of HCC with ablation. Fifteen relevant studies were included and further analyzed. Among them, three clinical studies suggested that HIF-1α might serve as a crucial role in the RAF treatment of HCC or the local recurrence of HCC after RFA. The remainder included experimental studies demonstrated that HIF-1, 2α might target the different molecules (e.g., BNIP3, CA-IX, and arginase-1) and signaling cascades (e.g., VEGFA/EphA2 pathway), constituting a complex network that promoted HCC invasion and metastasis after ablation. Currently, the inhibitors of HIF have been developed, providing important proof of targeting HIF for the prevention of HCC recurrence after IRFA and HIFU ablation. Further confirmation by prospective clinical and in-depth experimental studies is still warranted to illustrate the effects of HIF in HCC recurrence followed ablation treatment in the future.
Collapse
Affiliation(s)
- Chunying Xiao
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Sheng Liu
- Department of Hepatobiliary Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ge Ge
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Hao Jiang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Liezhi Wang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Qi Chen
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Chong Jin
- Department of General Surgery, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Jinggang Mo
- Department of General Surgery, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Jin Li
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Kunpeng Wang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Qianqian Zhang
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianyu Zhou
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| |
Collapse
|
10
|
Yu L, Cheng M, Liu J, Ye X, Wei Z, Xu J, Xie Q, Liang J. Crosstalk between microwave ablation and ferroptosis: The next hot topic? Front Oncol 2023; 13:1099731. [PMID: 36712497 PMCID: PMC9880492 DOI: 10.3389/fonc.2023.1099731] [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: 11/16/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Microwave ablation has been one form of thermal ablation in treatments for many tumors, which can locally control unresectable tumors. Ferroptosis is iron-dependent cell death caused by the cumulative reactive oxygen species and lipid peroxidation products. Recently, increasing evidence has shown that ferroptosis might play a vital role in MWA-induced tumor suppression. In this article, we briefly illustrate the concept of ferroptosis, the related signal pathways and inducers, the basic principle of microwave ablation in killing tumors, and the key molecules released after microwave ablation. Then, we describe the cross-talking molecules between microwave ablation and ferroptosis, and discussed the potential mechanism of microwave ablation-induced ferroptosis. This review explores the therapeutic target of ferroptosis in enhancing the systemic antitumor effect after microwave ablation, providing theoretical support in combinational microwave ablation with pro-ferroptosis therapy.
Collapse
Affiliation(s)
- Lu Yu
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Min Cheng
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jie Liu
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China,School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Xin Ye
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zhigang Wei
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jiamei Xu
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Qi Xie
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China,*Correspondence: Qi Xie, ; Jing Liang,
| | - Jing Liang
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China,*Correspondence: Qi Xie, ; Jing Liang,
| |
Collapse
|
11
|
Wei F, Guo R, Yan Y, Lin R, Chen J, Lin Z. Investigation of the efficacy and safety of cryoablation and intra-arterial PD-1 inhibitor in patients with advanced disease not responding to checkpoint inhibitors: An exploratory study. Front Immunol 2022; 13:990224. [PMID: 36211329 PMCID: PMC9537743 DOI: 10.3389/fimmu.2022.990224] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/09/2022] [Indexed: 11/26/2022] Open
Abstract
Objective To explore the effectiveness of cryoablation combined with arterial perfusion with programmed cell death protein 1 inhibitors in overcoming immune resistance in advanced solid cancers. Methods In this pilot retrospective study, nine patients with solid cancers were treated with tumour cryoablation and arterial perfusion with programmed cell death protein 1 inhibitors, which had previously proven ineffective. The CIBERSORT software was used to estimate the levels of tumour-infiltrating immune cells in the challenged tumour. Changes in the levels of circulating T cells were assessed using flow cytometry. The primary endpoints were disease control and objective response rates, and the secondary endpoint was safety. Results The nine patients with advanced solid tumours received cryoablation combined with arterial perfusion with programmed cell death protein 1 inhibitors between June and December 2021. The median follow-up time was 5.8 months. We recorded an objective response rate in two patients (22.22%). The best overall responses were partial responses in two patients (22.22%) and one case (11.11%) of stable disease, while six patients (66.67%) presented progressive disease. However, the median overall survival time was not reached. The median progression-free survival was 2.4 months. Treatment-related severe adverse events included one case of abdominal infection and one case of upper gastrointestinal bleeding, which were cured after the intervention. The CIBERSORT software confirmed the importance of cryoablation in regulating tumour-infiltrating immune cells. Thus, macrophage polarisation from the M2 to the M1 phenotype in the challenged tumour and a gradual increase in the levels of circulating CD4+ T cells were observed after administration of the combination therapy. Conclusion Cryoablation combined with arterial perfusion with programmed cell death protein 1 inhibitors has the potential efficacy and safety to overcome immune resistance in patients with advanced solid cancers. The combination therapy leads to macrophage polarisation from the M2 to the M1 phenotype in the challenged tumour to enhance antitumour immunity.
Collapse
Affiliation(s)
- Fuqun Wei
- The Department of Interventional Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Rui Guo
- The Department of Interventional Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Yuan Yan
- The Department of Interventional Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Ruixiang Lin
- The Department of Interventional Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jin Chen
- The Department of Interventional Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Zhengyu Lin
- The Department of Interventional Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, Fuzhou, Fujian, China
- *Correspondence: Zhengyu Lin,
| |
Collapse
|
12
|
Hsu FC, Lee HL, Chen YJ, Shen YA, Tsai YC, Wu MH, Kuo CC, Lu LS, Yeh SD, Huang WS, Shen CN, Chiou JF. A Few-Shot Learning Approach Assists in the Prognosis Prediction of Magnetic Resonance-Guided Focused Ultrasound for the Local Control of Bone Metastatic Lesions. Cancers (Basel) 2022; 14:445. [PMID: 35053608 PMCID: PMC8773927 DOI: 10.3390/cancers14020445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 01/05/2022] [Accepted: 01/13/2022] [Indexed: 11/24/2022] Open
Abstract
Magnetic resonance-guided focused ultrasound surgery (MRgFUS) constitutes a noninvasive treatment strategy to ablate deep-seated bone metastases. However, limited evidence suggests that, although cytokines are influenced by thermal necrosis, there is still no cytokine threshold for clinical responses. A prediction model to approximate the postablation immune status on the basis of circulating cytokine activation is thus needed. IL-6 and IP-10, which are proinflammatory cytokines, decreased significantly during the acute phase. Wound-healing cytokines such as VEGF and PDGF increased after ablation, but the increase was not statistically significant. In this phase, IL-6, IL-13, IP-10, and eotaxin expression levels diminished the ongoing inflammatory progression in the treated sites. These cytokine changes also correlated with the response rate of primary tumor control after acute periods. The few-shot learning algorithm was applied to test the correlation between cytokine levels and local control (p = 0.036). The best-fitted model included IL-6, IL-13, IP-10, and eotaxin as cytokine parameters from the few-shot selection, and had an accuracy of 85.2%, sensitivity of 88.6%, and AUC of 0.95. The acceptable usage of this model may help predict the acute-phase prognosis of a patient with painful bone metastasis who underwent local MRgFUS. The application of machine learning in bone metastasis is equivalent or better than the current logistic regression.
Collapse
Affiliation(s)
- Fang-Chi Hsu
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan;
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Hsin-Lun Lee
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan; (C.-C.K.); (L.-S.L.)
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Yin-Ju Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan;
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Yao-An Shen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yi-Chieh Tsai
- Department of Radiation Oncology, Shuang Ho Hospital, Taipei Medical University, Taipei 110, Taiwan;
| | - Meng-Huang Wu
- Department of Orthopedics, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Department of Orthopaedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Chia-Chun Kuo
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan; (C.-C.K.); (L.-S.L.)
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei 110, Taiwan
- Department of Radiation Oncology, Wanfang Hospital, Taipei Medical University, Taipei 110, Taiwan
| | - Long-Sheng Lu
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan; (C.-C.K.); (L.-S.L.)
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan;
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- International Ph.D. Program for Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Shauh-Der Yeh
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Urology, Taipei Medical University Hospital, Taipei 110, Taiwan
- Cancer Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Wen-Sheng Huang
- Department of Nuclear Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan;
| | - Chia-Ning Shen
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan;
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Jeng-Fong Chiou
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan;
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan; (C.-C.K.); (L.-S.L.)
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
13
|
Li H, Chen P, Wang M, Wang W, Li F, Han X, Ren J, Duan X. Liposome quercetin enhances the ablation effects of microwave ablation in treating the rabbit VX2 liver tumor model. Int J Hyperthermia 2022; 39:162-172. [PMID: 35000534 DOI: 10.1080/02656736.2021.2023767] [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: 08/22/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVE This study aimed to investigate whether liposomal quercetin (LQ) could enhance the effects of microwave ablation (MVA) in treating the rabbit VX2 liver tumor model. METHODS Rabbits with VX2 liver tumors were randomly divided into three groups: intravenous LQ group (LQ group), MWA group and LQ combined with MWA (LQ + MWA) group. Five rabbits were randomly selected and sacrificed from each group at 12 h and on days 3, 7 and 14 of the operation. The tumor samples were detected and quantified by immunohistochemistry, Western blot, and reverse transcription polymerase chain reaction (RT-PCR). RESULTS For up to 7 days, the coagulation necrosis volume (CV) of the LQ + MWA group was larger than that of MWA and LQ groups (p < 0.05). Fourteen days after the operation, the total tumor volume of the LQ + MWA group was smaller than that of the LQ group and the MWA group (p < 0.05). The survival time of the LQ + MWA group was significantly longer than that of the MWA and LQ groups (p < 0.01). Heat shock protein 70 (HSP70), hypoxia inducible factor-1 α (HIF-1 α), vascular endothelial growth factor (VEGF), tumor microvessel density (MVD) were lower in the LQ + MWA group than the MWA and LQ groups at 12 h, on days 3 and 7. At hour 12 and on days 3 and 7, HSP70 mRNA and HIF-1α mRNA expression of MWA group were significantly higher than that of the LQ and LQ + MWA groups (p < 0.001). At 12 h, and on days 3 and 7, apoptotic rate of tumor cells in LQ + MWA group was higher than that of the MWA and LQ groups (p < 0.05). At 12 h and on days 3, 7 and 14, the proliferation index of tumor cells in residual tumor in LQ + MWA group was lower than that in the MWA and LQ groups (p < 0.05). CONCLUSION Preoperative infusion of LQ can significantly enhance the MWA effects of liver VX2 tumor, inhibit the excessive proliferation of residual tumor and angiogenesis, and decrease metastasis and prolong the survival period of experimental animals.
Collapse
Affiliation(s)
- Hao Li
- Department of Interventional Radiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Pengfei Chen
- Department of Interventional Radiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Manzhou Wang
- Department of Interventional Radiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Wenhui Wang
- Department of Interventional Radiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Fangzheng Li
- Department of Interventional Radiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Jianzhuang Ren
- Department of Interventional Radiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Xuhua Duan
- Department of Interventional Radiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
14
|
Yu KH, Hung HY. Synthetic strategy and structure-activity relationship (SAR) studies of 3-(5'-hydroxymethyl-2'-furyl)-1-benzyl indazole (YC-1, Lificiguat): a review. RSC Adv 2021; 12:251-264. [PMID: 35424505 PMCID: PMC8978903 DOI: 10.1039/d1ra08120a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/14/2021] [Indexed: 01/04/2023] Open
Abstract
Since 1994, YC-1 (Lificiguat, 3-(5′-hydroxymethyl-2′-furyl)-1-benzylindazole) has been synthesized, and many targets for special bioactivities have been explored, such as stimulation of platelet-soluble guanylate cyclase, indirect elevation of platelet cGMP levels, and inhibition of hypoxia-inducible factor-1 (HIF-1) and NF-κB. Recently, Riociguat®, the first soluble guanylate cyclase (sGC) stimulator drug used to treat pulmonary hypertension and pulmonary arterial hypertension, was derived from the YC-1 structure. In this review, we aim to highlight the synthesis and structure–activity relationships in the development of YC-1 analogs and their possible indications. Since 1994, YC-1 (Lificiguat) has been synthesized, and many targets for special bioactivities have been explored, such as stimulation of platelet-soluble guanylate cyclase, indirect elevation of platelet cGMP levels, and inhibition of HIF-1 and NF-κB.![]()
Collapse
Affiliation(s)
- Ko-Hua Yu
- School of Pharmacy College of Medicine, National Cheng Kung University Tainan 701 Taiwan
| | - Hsin-Yi Hung
- School of Pharmacy College of Medicine, National Cheng Kung University Tainan 701 Taiwan
| |
Collapse
|
15
|
Chen Y, Bei J, Liu M, Huang J, Xie L, Huang W, Cai M, Guo Y, Lin L, Zhu K. Sublethal heat stress-induced O-GlcNAcylation coordinates the Warburg effect to promote hepatocellular carcinoma recurrence and metastasis after thermal ablation. Cancer Lett 2021; 518:23-34. [PMID: 34126196 DOI: 10.1016/j.canlet.2021.06.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/15/2021] [Accepted: 06/05/2021] [Indexed: 02/07/2023]
Abstract
The malignant transformation of residual hepatocellular carcinoma (HCC) cells after thermal ablation is considered as the main factor promoting postoperative HCC progression, which greatly limits the improvement of long-term survival, and at present there is no effective targeted therapeutic strategies. The Warburg effect is a metabolic feature correlated highly with malignant transformation (e.g. epithelial-to-mesenchymal transition [EMT]). Here, we showed that sublethal heat stress triggered a stronger Warburg effect of HCC cells, which contributed to the thermotolerance and invasion of HCC cells. Sublethal heat stress-induced O-GlcNAcylation was involved in this process. Such enhanced Warburg effect in HCC cells may be eliminated through O-GlcNAcylation inhibition, resulting in impaired thermotolerance and EMT, and thereby preventing tumor recurrence and metastasis of HCC-bearing mice after insufficient thermal ablation. Finally, we present evidence that sublethal heat stress-induced O-GlcNAcylation regulates the Warburg effect in HCC cells by promoting hypoxia-inducible factor 1α (HIF-1α) stability. In conclusion, the present study suggests that O-GlcNAcylation coordinates the Warburg effect to promote HCC progression after thermal ablation, which may serve as a novel potential target for controlling postoperative HCC recurrence and metastasis.
Collapse
Affiliation(s)
- Ye Chen
- 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
| | - Jiaxin Bei
- 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
| | - Mingyu Liu
- 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
| | - Jingjun Huang
- 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
| | - Lulu Xie
- 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
| | - Wensou Huang
- 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
| | - Mingyue Cai
- 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
| | - Yongjian Guo
- 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
|
16
|
Timmer FE, Geboers B, Nieuwenhuizen S, Schouten EA, Dijkstra M, de Vries JJ, van den Tol MP, de Gruijl TD, Scheffer HJ, Meijerink MR. Locally Advanced Pancreatic Cancer: Percutaneous Management Using Ablation, Brachytherapy, Intra-arterial Chemotherapy, and Intra-tumoral Immunotherapy. Curr Oncol Rep 2021; 23:68. [PMID: 33864144 PMCID: PMC8052234 DOI: 10.1007/s11912-021-01057-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive neoplasms, bearing a terrible prognosis. Stage III tumors, also known as locally advanced pancreatic cancer (LAPC), are unresectable, and current palliative chemotherapy regimens have only modestly improved survival in these patients. At this stage of disease, interventional techniques may be of value and further prolong life. The aim of this review was to explore current literature on locoregional percutaneous management for LAPC. RECENT FINDINGS Locoregional percutaneous interventional techniques such as ablation, brachytherapy, and intra-arterial chemotherapy possess cytoreductive abilities and have the potential to increase survival. In addition, recent research demonstrates the immunomodulatory capacities of these treatments. This immune response may be leveraged by combining the interventional techniques with intra-tumoral immunotherapy, possibly creating a durable anti-tumor effect. This multimodality treatment approach is currently being examined in several ongoing clinical trials. The use of certain interventional techniques appears to improve survival in LAPC patients and may work synergistically when combined with immunotherapy. However, definitive conclusions can only be made when large prospective (randomized controlled) trials confirm these results.
Collapse
Affiliation(s)
- Florentine E.F. Timmer
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Bart Geboers
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Sanne Nieuwenhuizen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Evelien A.C. Schouten
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Madelon Dijkstra
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Jan J.J. de Vries
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - M. Petrousjka van den Tol
- Department of Surgical Oncology, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Tanja D. de Gruijl
- Department of Medical Oncology, Amsterdam UMC (location VUmc)-Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Hester J. Scheffer
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Martijn R. Meijerink
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
17
|
Jiang AN, Liu JT, Zhao K, Wu H, Wang S, Yan K, Yang W. Specific Inhibitor of Matrix Metalloproteinase Decreases Tumor Invasiveness After Radiofrequency Ablation in Liver Tumor Animal Model. Front Oncol 2020; 10:561805. [PMID: 33330030 PMCID: PMC7709861 DOI: 10.3389/fonc.2020.561805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/19/2020] [Indexed: 12/26/2022] Open
Abstract
Objective To determine whether the specific inhibitor of matrix metalloproteinase (MMP)-batimastat (BB-94)-could decrease the progression of liver tumor after radiofrequency ablation (RFA) and achieve better therapeutic efficacy in an animal model. Methods In vitro experiments, the proliferation of H22 liver tumor cells was detected by CCK 8 assay and cell migration was detected by Transwell method. In vivo experiments, H22 murine liver tumors were used. First, 32 mice with one tumor were randomized into four groups (n = 8 each group): control (PBS only), RFA alone (65°C, 5 min), BB-94 (30 mg/kg), RFA+BB-94. The growth rate of the residual tumor and the end point survival were calculated and the pathologic changes were evaluated. Secondly, a total of 48 tumors in 24 animals (paired tumors) were randomized into three groups (n = 8 each group): control, RFA alone, RFA+BB-94. Each mouse was implanted with two tumors subcutaneously, one tumor was treated by RFA and the other was evaluated for distant metastasis after applying BB-94. Results In vitro, the proliferation assay demonstrated higher proliferation ability after heat treatment (0.82 ± 0.07 vs 1.27 ± 0.08, P = 0.008), and it could be inhibited by BB-94 (1.27 ± 0.08 vs 0.67 ± 0.06, P = 0.001). In the cell migration assay, the H22 cells demonstrated enhanced tumor invasiveness in the heat group than the control group (33.7 ± 2.1 vs 19.7 ± 4.9, P = 0.011). And it could be significantly suppressed after BB-94 incubation (33.7 ± 2.1 vs 23.0 ± 4.6, P = 0.009). With one tumor animal, the growth rate of the residual tumor in the BB-94+RFA group was slower than that in the RFA alone group (P = 0.003). And combination of BB-94 could significantly prolong the survival of the mice (40.3 ± 1.4d vs 47.1 ± 1.3d, P = 0.002). The expression of CD31 and VEGF at the coagulation margin were decreased after combined with BB-94. With two tumors animal, the growth of metastasis tumor in the BB-94+RFA group was slower than that in the RFA group (P < 0.001). Conclusion BB-94 combined with RFA reduced the invasiveness of the liver tumor and improved the end-point survival. Our data suggested that targeting the MMP process with the specific inhibition could help to increase overall ablation efficacy.
Collapse
Affiliation(s)
- An-Na Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jing-Tao Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pharmacy, Peking University Cancer Hospital & Institute, Beijing, China
| | - Kun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hao Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Song Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Kun Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wei Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
18
|
Modelling of combination therapy using implantable anticancer drug delivery with thermal ablation in solid tumor. Sci Rep 2020; 10:19366. [PMID: 33168846 PMCID: PMC7653950 DOI: 10.1038/s41598-020-76123-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 10/22/2020] [Indexed: 12/20/2022] Open
Abstract
Local implantable drug delivery system (IDDS) can be used as an effective adjunctive therapy for solid tumor following thermal ablation for destroying the residual cancer cells and preventing the tumor recurrence. In this paper, we develop comprehensive mathematical pharmacokinetic/pharmacodynamic (PK/PD) models for combination therapy using implantable drug delivery system following thermal ablation inside solid tumors with the help of molecular communication paradigm. In this model, doxorubicin (DOX)-loaded implant (act as a transmitter) is assumed to be inserted inside solid tumor (acts as a channel) after thermal ablation. Using this model, we can predict the extracellular and intracellular concentration of both free and bound drugs. Also, Impact of the anticancer drug on both cancer and normal cells is evaluated using a pharmacodynamic (PD) model that depends on both the spatiotemporal intracellular concentration as well as characteristics of anticancer drug and cells. Accuracy and validity of the proposed drug transport model is verified with published experimental data in the literature. The results show that this combination therapy results in high therapeutic efficacy with negligible toxicity effect on the normal tissue. The proposed model can help in optimize development of this combination treatment for solid tumors, particularly, the design parameters of the implant.
Collapse
|
19
|
Chen Y, Wang K, Di J, Guan C, Wang S, Li Q, Qu Y. Mutation of the BAG-1 domain decreases its protective effect against hypoxia/reoxygenation by regulating HSP70 and the PI3K/AKT signalling pathway in SY-SH5Y cells. Brain Res 2020; 1751:147192. [PMID: 33152339 DOI: 10.1016/j.brainres.2020.147192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 09/30/2020] [Accepted: 10/28/2020] [Indexed: 10/23/2022]
Abstract
BCL-2-associated athanogene-1 (BAG-1) is a multifunctional protein that was first identified as a binding partner of BCL-2. Our previous results indicated that BAG-1 large (BAG-1L) overexpression significantly increases cell viability and decreases apoptosis by upregulating HSP70 and p-AKT in response to hypoxia/reoxygenation in SY-SH5Y cells. However, the functional domain of BAG-1L that exerts these protective effects against hypoxic damage has not been identified. In this study, we examined changes in HSP70 and p-AKT protein levels in SH-SY5Y cells with or without BAG-1L domain mutation after six hours of hypoxia/reoxygenation treatment. The BAG-1 domain mutant (BAG-1MUT) attenuated neuronal viability and proliferation while enhancing apoptosis after hypoxia/reoxygenation, which was achieved in part by inhibiting the HSP70 and p-AKT signalling pathways. This evidence illustrates that the BAG-1 domain plays a key role in protecting cells from hypoxia/reoxygenation injury.
Collapse
Affiliation(s)
- Ying Chen
- School of Nursing, Medical College of Qingdao University, Qingdao 26600, Shandong, China; Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Keke Wang
- Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Jie Di
- School of Nursing, Medical College of Qingdao University, Qingdao 26600, Shandong, China; Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Chun Guan
- Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Sumei Wang
- Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Qingshu Li
- Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China.
| | - Yan Qu
- Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| |
Collapse
|
20
|
Li Z, Jiang M, Zhang T, Liu S. GAS6-AS2 Promotes Hepatocellular Carcinoma via miR-3619-5p/ARL2 Axis Under Insufficient Radiofrequency Ablation Condition. Cancer Biother Radiopharm 2020; 36:879-887. [PMID: 32799553 DOI: 10.1089/cbr.2019.3541] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a common malignancy worldwide. Radiofrequency ablation (RFA) is applied for treating HCC; however, insufficient RFA promotes HCC development and accelerates HCC recurrence. Therefore, the molecular functions underlying this process have gradually attracted attention. Aim of the study: We sought to examine whether GAS6-AS2 (also known as GAS6-DT: growth arrest specific 6 divergent transcript) played a role in the development of HCC after insufficient RFA. Methods: The in vitro model was established by heating Huh7 and MHCC97 cells in water bath at 47°C, named as Huh7-H and MHCC97-H. Colony formation, transwell and western blot assays were conducted for functional analysis. Results: GAS6-AS2 was upregulated in Huh7-H and MHCC97-H cells relative to Huh7 and MHCC97 cells. GAS6-AS2 deficiency hampered cell proliferation, migration, invasion, epithelial-mesenchymal transition, and stemness in Huh7-H and MHCC97-H cells. Moreover, microRNA-3619-5p (miR-3619-5p) combined with GAS6-AS2 and ARL2 (ADP ribosylation factor-like GTPase 2) was the target gene of miR-3619-5p. GAS6-AS2 served as the competing endogenous RNA (ceRNA) of ARL2 via absorbing miR-3619-5p. Conclusion: On the whole, present study uncovered a novel ceRNA mechanism of GAS6-AS2/miR-3619-5p/ARL2 in HCC after insufficient RFA, which might shed a new insight into treatment of HCC after insufficient RFA.
Collapse
Affiliation(s)
- Zilin Li
- Invasive Technology Department, Hubei Cancer Hospital, Wuhan, China
| | - Manhong Jiang
- Oncology Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Ting Zhang
- Chest Radiotherapy Department 1, Hubei Cancer Hospital, Wuhan, China
| | - Suhua Liu
- Anesthesiology Department, Hubei Rongjun Hospital, Wuhan, China
| |
Collapse
|
21
|
Kong J, Yao C, Ding X, Dong S, Wu S, Sun W, Zheng L. ATPase Inhibitory Factor 1 Promotes Hepatocellular Carcinoma Progression After Insufficient Radiofrequency Ablation, and Attenuates Cell Sensitivity to Sorafenib Therapy. Front Oncol 2020; 10:1080. [PMID: 32670888 PMCID: PMC7330926 DOI: 10.3389/fonc.2020.01080] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 05/29/2020] [Indexed: 12/20/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) and angiogenesis is involved in tumor progression after radiofrequency ablation (RFA). ATPase inhibitory factor 1 (IF1) is a bad predictor of prognosis. Sorafenib inhibited EMT of hepatocellular carcinoma (HCC) after RFA. Whether IF1 promotes the EMT and angiogenesis of HCC and attenuates the effect of sorafenib after insufficient RFA is investigated. In this study, higher expression of IF1 was found in residual tumor after insufficient RFA. Hep3B or Huh7 cells after insufficient RFA were designated as Hep3B-H or Huh7-H cells in vitro. Hep3B-H or Huh7-H cells exhibited enhanced capacities of colony formation, migration, and increased expression of EMT associated markers and IF1 compared with Hep3B or Huh7 cells. IF1 knockdown in Hep3B-H or Huh7-H cells decreased the colony formation and migratory capacity, and IF1 overexpression in Hep3B or Huh7 cells increased these capacities. IF1 in HCC cells directly and indirectly affected angiogenesis of TAECs after insufficient RFA. IF1 promoted HCC cells growth and metastasis after insufficient RFA. IF1 increased HCC cells resistance after insufficient RFA to sorafenib. Higher IF1 expression indicated poor disease survival in HCC patients after sorafenib therapy. NF-κB activation induced by IF1 attenuated the effect of sorafenib on HCC cells after insufficient RFA. Our results demonstrated that IF1 promotes the EMT and angiogenesis, and attenuates HCC cell sensitivity to sorafenib after insufficient RFA through NF-κB signal pathway.
Collapse
Affiliation(s)
- Jian Kong
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Changyu Yao
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xuemei Ding
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Shuying Dong
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Shilun Wu
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wenbing Sun
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Lemin Zheng
- Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, School of Basic Medical Sciences, The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, Peking University Health Science Center, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Chen S, Yang S, Xu S, Dong S. Comparison between radiofrequency ablation and sublobar resections for the therapy of stage I non-small cell lung cancer: a meta-analysis. PeerJ 2020; 8:e9228. [PMID: 32509468 PMCID: PMC7246024 DOI: 10.7717/peerj.9228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/30/2020] [Indexed: 12/16/2022] Open
Abstract
Background Sublobar resection (SLR) and radiofrequency ablation (RFA) are the two minimally invasive procedures performed for treating stage I non-small cell lung cancer (NSCLC). This study aimed to compare SLR and RFA for the treatment of stage I NSCLC using the meta-analytical method. Methods We searched PubMed and Embase for articles published till December 2019 to evaluate the comparative studies and assess the survival and progression-free survival rates and postoperative complications (PROSPERO registration number: CRD42018087587). A meta-analysis was performed by combining the outcomes of the reported incidences of short-term morbidity and long-term mortality. The fixed or random effects model was utilized to calculate the pooled odds ratios (OR) and the 95% confidence intervals. Results Four retrospective studies were considered in the course of this study. The studies included a total of 309 participants; 154 were assigned to the SLR group, and 155 were assigned to the RFA group. Moreover, there were statistically significant differences between the one- and three-year survival rates and one- and three-year progression-free survival rates for the two groups, which were in favor of the SLR group. Among the post-surgical complications, pneumothorax and pleural effusion were more common for the SLR group, while cardiac abnormalities were prevalent in the RFA group. There was no difference in prevalence of hemoptysis between SLR and RFA groups, which might be attributed to the limited study sample size. Conclusion Considering the higher survival rates and disease control in the evaluated cases, surgical resection is the preferred treatment method for stage I NSCLC. RFA can be considered a valid alternative in patients not eligible for surgery and in high-risk patients as it is less invasive and requires shorter hospital stay.
Collapse
Affiliation(s)
- Shuang Chen
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Shize Yang
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Shun Xu
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Siyuan Dong
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
23
|
Wan J, Ling X, Rao Z, Peng B, Ding G. Independent prognostic value of HIF-1α expression in radiofrequency ablation of lung cancer. Oncol Lett 2020; 19:849-857. [PMID: 31897199 PMCID: PMC6924154 DOI: 10.3892/ol.2019.11130] [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: 02/01/2019] [Accepted: 09/20/2019] [Indexed: 01/11/2023] Open
Abstract
Radiofrequency ablation (RFA) is widely used in the treatment of lung cancer. Hypoxia-inducible factor-1α (HIF-1α) is a crucial transcription factor regulating oxygen homeostasis that is involved in tumor cell metastasis. The present study investigated the impact of HIF-1α expression and other factors, such as postoperative blood CD4+/CD8+ ratio, on the prognosis of patients with lung cancer who had received RFA treatment. A total of 80 patients with lung cancer were recruited between January 2011 and October 2016 at The Shenzhen People's Hospital. Lung cancer was confirmed following pathological or histological examination. All patients underwent RFA treatment. Patients were followed up for 6–66 months. HIF-1α expression in lung cancer tissues was assessed by immunohistochemistry. Multivariate survival analysis was performed using Cox proportional hazards model. The results demonstrated that HIF-1α level was low in 36 patients and overexpressed in 44 patients with lung cancer. Kaplan-Meier (KM) curve analysis demonstrated that the overall survival time of patients with high HIF-1α expression was significantly shorter compared with patients with low HIF-1α expression (P<0.05). Furthermore, the results from the KM model and log-rank test revealed that age, Union for International Cancer Control stage, primary or metastatic cancer, chemotherapy, postoperative blood CD4+/CD8+ ratio, Eastern Cooperative Oncology Group performance status and HIF-1α expression had significant effects on overall survival of patients with lung cancer. The results from Cox analysis demonstrated that high HIF-1α expression, advanced age, clinical staging and chemotherapy were independent risk factors for the prognosis of lung cancer following RFA treatment, and that high HIF-1α expression was associated with the increased risk (5.91-fold) of mortality. In conclusion, the present study demonstrated that HIF-1α expression was increased in lung cancer tissues and was associated with the prognosis of patients with lung cancer who were treated with RFA. These findings suggest that HIF-1α expression may be considered as a marker for evaluating the prognosis of these patients.
Collapse
Affiliation(s)
- Jun Wan
- Department of Thoracic Surgery, The Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Xiean Ling
- Department of Thoracic Surgery, The Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Zhanpeng Rao
- Department of Thoracic Surgery, The Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Bin Peng
- Department of Thoracic Surgery, The Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Guanggui Ding
- Department of Thoracic Surgery, The Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
24
|
Duan XH, Li H, Han XW, Ren JZ, Li FY, Ju SG, Chen PF, Kuang DL. Upregulation of IL-6 is involved in moderate hyperthermia induced proliferation and invasion of hepatocellular carcinoma cells. Eur J Pharmacol 2018; 833:230-236. [DOI: 10.1016/j.ejphar.2018.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/08/2018] [Accepted: 06/08/2018] [Indexed: 02/07/2023]
|
25
|
Impact of Interventional Oncology Therapies on Tumor Microenvironment and Strategies to Enhance Their Efficacy. AJR Am J Roentgenol 2018; 210:648-656. [PMID: 29364726 DOI: 10.2214/ajr.16.17677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE We provide a brief review of the tumor microenvironment, the impact of six interventional radiology treatments on the tumor microenvironment, and potential methods to improve treatment efficacy. CONCLUSION Interventional oncology plays a unique role in cancer therapy, contributing to both antitumorigenic and protumorigenic effects.
Collapse
|
26
|
Impact of Elevated Circulating Histones on Systemic Inflammation after Radiofrequency Ablation in Lung Cancer Patients. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6894832. [PMID: 29457035 PMCID: PMC5804403 DOI: 10.1155/2017/6894832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/26/2017] [Indexed: 11/17/2022]
Abstract
Background This study investigated the changes of circulating histones following radiofrequency ablation (RFA) in lung cancer patients and their impact on systemic inflammation. Methods Serial blood samples were obtained from a total of 65 primary and metastatic lung cancer patients undergoing RFA at 2 time points: pre-RFA and post-RFA within 48 h. Circulating histones, myeloperoxidase (MPO), and multiple inflammatory cytokines were measured. Moreover, the patient's sera were incubated overnight with human monocytic U937 cells in the presence or absence of anti-histone antibody, and cytokine production was evaluated. Results Compared to pre-RFA, there was a significant increase in circulating histones within 48 h after RFA, along with an elevation of MPO and several canonical inflammatory cytokines. Circulating histones were correlated with these inflammatory markers. Notably, compared to the sera obtained before RFA, the patients' post-RFA sera significantly stimulated cytokine production in the supernatant of U937 cells, which could be prevented by anti-histone antibody, thereby confirming a cause-effect relationship between circulating histones and systemic inflammation. Conclusions This study showed that circulating histones may serve as a marker indicating RFA-related systemic inflammation as well as represent a therapeutic target for resolution of inflammation.
Collapse
|
27
|
Duan X, Chen P, Han X, Ren J, Wang Z, Zhao G, Li H. The influence of liposomal quercetin on liver damage induced by microwave ablation. Sci Rep 2017; 7:12677. [PMID: 28978941 PMCID: PMC5627272 DOI: 10.1038/s41598-017-13010-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/14/2017] [Indexed: 12/31/2022] Open
Abstract
This study aimed to observe whether liposomal quercetin (LQ) can enhance the effect of microwave ablation (MWA) on hepatic parenchyma destruction. Forty-eight rabbits were randomly divided into three groups: LQ group, MWA group and LQ + MWA group. Serum and liver samples were collected. The coagulation volume (CV) of hepatic parenchyma, histopathological changes and liver function were compared. Hepatocyte apoptosis was examined through TUNEL. The expression of heat shock protein 70 (HSP70), hypoxia-inducible factor-1α (HIF-1α) and tumor necrosis factor-α (TNF-α) were analyzed. Compared with MWA group, the CV of coagulation necrosis in liver was significantly increased in LQ + MWA group. TUNEL results showed that the hepaocyte apoptosis was higher in LQ + MWA group than MWA group on 12 h, 24 h and 3 d, respectively. HSP70 and HIF-1α expression in both MWA group and LQ + MWA group were increased at 12 and 24 hours, peaked on day3 and dropped on day7. Compared with MWA group, HSP70 and HIF-1α expression were lower in LQ + MWA group. On the contrary, TNF-α expression was decreased in MWA group and LQ + MWA group compared with LQ group. In conclusion, LQ increased hepatocyte apoptosis and MWA-induced hepatic parenchyma destruction through suppressing HSP70 and HIF-1α expression in liver surrounding ablation zone and increasing TNF-α expression.
Collapse
Affiliation(s)
- Xuhua Duan
- Department of Radiology, The First Affiliated Hospital, Zhengzhou University, No. 1, East Jian She Road, Zhengzhou, 450052, Henan Province, People's Republic of China
| | | | - Xinwei Han
- Department of Radiology, The First Affiliated Hospital, Zhengzhou University, No. 1, East Jian She Road, Zhengzhou, 450052, Henan Province, People's Republic of China.
| | - Jianzhuang Ren
- Department of Radiology, The First Affiliated Hospital, Zhengzhou University, No. 1, East Jian She Road, Zhengzhou, 450052, Henan Province, People's Republic of China.
| | | | | | | |
Collapse
|
28
|
Lee MR, Lin C, Lu CC, Kuo SC, Tsao JW, Juan YN, Chiu HY, Lee FY, Yang JS, Tsai FJ. YC-1 induces G 0/G 1 phase arrest and mitochondria-dependent apoptosis in cisplatin-resistant human oral cancer CAR cells. Biomedicine (Taipei) 2017; 7:12. [PMID: 28612710 PMCID: PMC5479426 DOI: 10.1051/bmdcn/2017070205] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/02/2017] [Indexed: 12/15/2022] Open
Abstract
Oral cancer is a serious and fatal disease. Cisplatin is the first line of chemotherapeutic agent for oral cancer therapy. However, the development of drug resistance and severe side effects cause tremendous problems clinically. In this study, we investigated the pharmacologic mechanisms of YC-1 on cisplatin-resistant human oral cancer cell line, CAR. Our results indicated that YC-1 induced a concentration-dependent and time-dependent decrease in viability of CAR cells analyzed by MTT assay. Real-time image analysis of CAR cells by IncuCyte™ Kinetic Live Cell Imaging System demonstrated that YC-1 inhibited cell proliferation and reduced cell confluence in a time-dependent manner. Results from flow cytometric analysis revealed that YC-1 promoted G0/G1 phase arrest and provoked apoptosis in CAR cells. The effects of cell cycle arrest by YC-1 were further supported by up-regulation of p21 and down-regulation of cyclin A, D, E and CDK2 protein levels. TUNEL staining showed that YC-1 caused DNA fragmentation, a late stage feature of apoptosis. In addition, YC-1 increased the activities of caspase-9 and caspase-3, disrupted the mitochondrial membrane potential (AYm) and stimulated ROS production in CAR cells. The protein levels of cytochrome c, Bax and Bak were elevated while Bcl-2 protein expression was attenuated in YC-1-treated CAR cells. In summary, YC-1 suppressed the viability of cisplatin-resistant CAR cells through inhibiting cell proliferation, arresting cell cycle at G0/G1 phase and triggering mitochondria-mediated apoptosis. Our results provide evidences to support the potentially therapeutic application of YC-1 on fighting against drug resistant oral cancer in the future.
Collapse
Affiliation(s)
- Miau-Rong Lee
- Department of Biochemistry, China Medical University, Taichung 404, Taiwan
| | - Chingju Lin
- Department of Physiology, China Medical University, Taichung 404, Taiwan
| | - Chi-Cheng Lu
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan - Department of Pharmacy, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan
| | - Sheng-Chu Kuo
- Chinese Medicinal Research and Development Center, China Medical University Hospital, China Medical University, Taichung 404, Taiwan - School of Pharmacy, China Medical University, Taichung 404, Taiwan
| | - Je-Wei Tsao
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Yu-Ning Juan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Hong-Yi Chiu
- Department of Pharmacy, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan
| | - Fang-Yu Lee
- Yung-Shin Pharmaceutical Industry Co., Ltd., Tachia, Taichung 437, Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Fuu-Jen Tsai
- Genetics Center, Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan - School of Chinese Medicine, China Medical University, Taichung 404, Taiwan - Department of Medical Genetics, China Medical University Hospital, Taichung 404, Taiwan
| |
Collapse
|
29
|
Sheng SR, Wu JS, Tang YL, Liang XH. Long noncoding RNAs: emerging regulators of tumor angiogenesis. Future Oncol 2017; 13:1551-1562. [PMID: 28513194 DOI: 10.2217/fon-2017-0149] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) participate in multiple biological processes especially human diseases, of which, tumor seems to be one of the most significant. Angiogenesis has been deemed to have a pivotal role in a series of tumor biological behaviors in tumorigenesis, progression and prognosis. Emerging evidences suggested that lncRNAs are involved in tumor angiogenesis and lncRNAs have already been verified to be potential biomarkers and promising therapeutic targets. This review summarized emerging angiogenesis-related lncRNAs, discussed their mechanisms interacting with cytokines, cancer stem cells, miRNAs and tumor hypoxia microenvironment, and demonstrated if lncRNAs could be new candidate targets of antiangiogenesis therapy.
Collapse
Affiliation(s)
- Su-Rui Sheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China.,Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China
| | - Jia-Shun Wu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China.,Department of Oral Pathology, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China.,Department of Oral Pathology, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China.,Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China
| |
Collapse
|