1
|
Chen C, Wang F, Cheng C, Li H, Fan Y, Jia L. Cancer-associated Fibroblasts-derived Exosomes with HOXD11 Overexpression Promote Ovarian Cancer Cell Angiogenesis Via FN1. Reprod Sci 2024:10.1007/s43032-024-01716-3. [PMID: 39394547 DOI: 10.1007/s43032-024-01716-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024]
Abstract
Cancer-associated fibroblasts (CAFs) represent a critical stromal component of metastatic niche and promote metastasis in patients with ovarian cancer (OC). Here, we try to further understand the mechanism by which CAFs-derived exosomes (CAFs-Exo) promoted angiogenesis in OC. We intersected differentially expressed genes in OC cells after CAFs-Exo treatment in the GSE147610 dataset with a list of transcription factors to identify homeobox protein hox-D11 (HOXD11) as a possible cargo of CAFs-Exo. HOXD11 encapsulated by CAFs-Exo enhanced colony formation, migration, and invasion of OC cells. HOXD11 bound to the promoter of fibronectin (FN1) and promoted its transcription. HOXD11 knockdown from CAFs-Exo significantly repressed the VEGF and CD31 protein expression and tube formation, viability, and migration of human umbilical vein endothelial cells (HUVEC) and slowed angiogenesis and tumor growth in mice. Furthermore, we found that overexpression of FN1 increased the expression of angiogenic factors and activity of HUVEC in the presence of HOXD11 knockdown. These results verify the significant contribution of CAFs-Exo to angiogenesis in OC, which could be partially due to the promotion of FN1 mediated by HOXD11.
Collapse
Affiliation(s)
- Chunfei Chen
- Department of Gynaecology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, P.R. China
| | - Fahui Wang
- Department of Gynaecology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, P.R. China
| | - Chunling Cheng
- Department of Gynaecology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, P.R. China
| | - Hongxin Li
- Department of Gynaecology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, P.R. China
| | - Yadan Fan
- Department of Gynaecology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, P.R. China
| | - Liping Jia
- Department of Gynaecology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, P.R. China.
| |
Collapse
|
2
|
Ran Y, Han S, Gao D, Chen X, Liu C. Interference of FZD2 suppresses proliferation, vasculogenic mimicry and stemness in glioma cells via blocking the Notch/NF‑κB signaling pathway. Exp Ther Med 2024; 28:373. [PMID: 39091630 PMCID: PMC11292164 DOI: 10.3892/etm.2024.12662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/22/2024] [Indexed: 08/04/2024] Open
Abstract
Frizzled family protein 2 (FZD2) is widely associated with tumor development and metastasis. The present study aimed to gain an insight into the role and regulatory mechanism of FZD2 in glioma. The expression level of FZD2 in normal astrocyte and glioma cells was determined by reverse transcription-quantitative PCR and western blotting, and cell transfection was conducted for FZD2 expression knockdown. Malignant behaviors including cell proliferation, migration and invasion, vasculogenic mimicry (VM) and cell stemness were determined using Cell Counting Kit-8, 5-Ethynyl-2'-deoxyuridine (EdU) staining, colony formation, wound healing, Transwell, 3D culturing and sphere formation assays. The expression levels of proteins related to stemness, epithelial-mesenchymal transition (EMT) and Notch/NF-κB signaling were measured by western blotting. Then, the Notch agonist, Jagged-1 (JAG), was adopted for rescue experiments. The results demonstrated that FZD2 was highly expressed in glioma cells. Interference of FZD2 expression suppressed the proliferation of glioma cells, as evidenced by the reduced cell viability and the number of EdU+ cells and colonies. Meanwhile, the reduced sphere formation ability and decreased protein expression of Nanog, Sox2 and Oct4 following FZD2 knockdown confirmed that FZD2 repressed cell stemness in glioma. Additionally, FZD2 knockdown suppressed the migration, invasion, EMT and VM formation capabilities of glioma cells, and also blocked the Notch/NF-κB signaling pathway. Furthermore, activation of Notch by JAG treatment partially reversed the aforementioned FZD2 knockdown-mediated changes in glioma cell malignant behaviors. In conclusion, FZD2 may contribute to glioma progression through activating the Notch/NF-κB signaling pathway, providing a plausible therapeutic target for the treatment of glioma.
Collapse
Affiliation(s)
- Yuge Ran
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Shuai Han
- Department of Medicine, Batai Biopharmaceutical Co., Ltd., Beijing 102600, P.R. China
| | - Dongxue Gao
- Proton Therapy Center, Cancer Hospital Chinese Academy of Medical Sciences, Langfang, Hebei 065000, P.R. China
| | - Xiaobo Chen
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Chan Liu
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| |
Collapse
|
3
|
Wang S, Miao J, Zhu P, Xu L. Co-delivery of Liposomal Ketoconazole and Bevacizumab for Synergistical Inhibition of Angiogenesis Against Endometrial Cancer. Mol Biotechnol 2024:10.1007/s12033-024-01227-1. [PMID: 39230827 DOI: 10.1007/s12033-024-01227-1] [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: 09/08/2023] [Accepted: 06/20/2024] [Indexed: 09/05/2024]
Abstract
In this study, we designed a novel formulation based on liposomes for the co-delivery of cancer-derived exosome inhibitor (ketoconazole, Keto) and angiogenesis inhibitor (bevacizumab, mAb). The designed Combo-Lipo formulation was systematically characterized, exhibiting a uniform average particle size of 100 nm, as well as excellent serum and long-term physical stabilities. The cell viability assay revealed that Combo-Lipo treatment significantly reduced the viability of cancer cells compared to free drugs. Moreover, liposomes effectively inhibited angiogenic mediators and reduced tumor immune suppressive factors. The Combo-Lipo formulation demonstrated potent downregulation of angiogenic factors and synergistic effects in suppressing their production. Furthermore, liposomes inhibited tumor-associated macrophages (TAMs), leading to decreased expression of tumor-promoting factors. Together, these findings highlighted the promising characteristics of Combo-Lipo as a therapeutic formulation, including optimal particle size, serum stability, and potent anti-cancer effects, as well as inhibition of angiogenic mediators and TAMs toward treating endometrial cancer.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of the First Obstetrics and Gynecology, Yantai Yuhuangding Hospital, Yantai, 264099, Shandong, China
| | - Jinglin Miao
- Department of the Third Obstetrics and Gynecology, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai, 264099, Shandong, China
| | - Ping Zhu
- Department of Reproductive Medicine, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai, 264099, Shandong, China.
| | - Li Xu
- Department of the Third Obstetrics and Gynecology, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai, 264099, Shandong, China.
| |
Collapse
|
4
|
Zhang J, Ouyang F, Gao A, Zeng T, Li M, Li H, Zhou W, Gao Q, Tang X, Zhang Q, Ran X, Tian G, Quan X, Tang Z, Zou J, Zeng Y, Long Y, Li Y. ESM1 enhances fatty acid synthesis and vascular mimicry in ovarian cancer by utilizing the PKM2-dependent warburg effect within the hypoxic tumor microenvironment. Mol Cancer 2024; 23:94. [PMID: 38720298 PMCID: PMC11077861 DOI: 10.1186/s12943-024-02009-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND The hypoxic tumor microenvironment is a key factor that promotes metabolic reprogramming and vascular mimicry (VM) in ovarian cancer (OC) patients. ESM1, a secreted protein, plays an important role in promoting proliferation and angiogenesis in OC. However, the role of ESM1 in metabolic reprogramming and VM in the hypoxic microenvironment in OC patients has not been determined. METHODS Liquid chromatography coupled with tandem MS was used to analyze CAOV3 and OV90 cells. Interactions between ESM1, PKM2, UBA2, and SUMO1 were detected by GST pull-down, Co-IP, and molecular docking. The effects of the ESM1-PKM2 axis on cell glucose metabolism were analyzed based on an ECAR experiment. The biological effects of the signaling axis on OC cells were detected by tubule formation, transwell assay, RT‒PCR, Western blot, immunofluorescence, and in vivo xenograft tumor experiments. RESULTS Our findings demonstrated that hypoxia induces the upregulation of ESM1 expression through the transcription of HIF-1α. ESM1 serves as a crucial mediator of the interaction between PKM2 and UBA2, facilitating the SUMOylation of PKM2 and the subsequent formation of PKM2 dimers. This process promotes the Warburg effect and facilitates the nuclear translocation of PKM2, ultimately leading to the phosphorylation of STAT3. These molecular events contribute to the promotion of ovarian cancer glycolysis and vasculogenic mimicry. Furthermore, our study revealed that Shikonin effectively inhibits the molecular interaction between ESM1 and PKM2, consequently preventing the formation of PKM2 dimers and thereby inhibiting ovarian cancer glycolysis, fatty acid synthesis and vasculogenic mimicry. CONCLUSION Our findings demonstrated that hypoxia increases ESM1 expression through the transcriptional regulation of HIF-1α to induce dimerization via PKM2 SUMOylation, which promotes the OC Warburg effect and VM.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Fan Ouyang
- Department of Cardiology, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Anbo Gao
- Department of Cardiology, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
- Clinical Research Institute, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tian Zeng
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ming Li
- Trauma Center, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Wenchao Zhou
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qing Gao
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xing Tang
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Qunfeng Zhang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaomin Ran
- Department of Gynecologic Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Gang Tian
- Department of Rehabilitation, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Xiyun Quan
- Department of Pathology, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Zhenzi Tang
- Department of Gynecologic Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Juan Zou
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yifei Zeng
- Department of Oncology, Shenzhen Luohu People's Hospital, Shenzhen, Guangdong, China.
| | - Yunzhu Long
- Department of Infectious Disease, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China.
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China.
| |
Collapse
|
5
|
Liu Y, Sun B, Lin Y, Deng H, Wang X, Xu C, Wang K, Yu N, Liu R, Han M. Lysyl Oxidase Promotes the Formation of Vasculogenic Mimicry in Gastric Cancer through PDGF-PDGFR Pathway. J Cancer 2024; 15:1816-1825. [PMID: 38434983 PMCID: PMC10905410 DOI: 10.7150/jca.92192] [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: 11/13/2023] [Accepted: 01/11/2024] [Indexed: 03/05/2024] Open
Abstract
Objective: Vasculogenic mimicry (VM) generates an important supplementary form of blood supply in cancer, which many factors regulate. However, the effect of lysyl oxidase (LOX) on VM formation is unclear. In this study, gastric cancer tissues and cells were used to investigate the role of LOX in the formation of VM. Materials and Methods: The samples were collected from 49 patients with a final diagnosis of gastric cancer. According to metastasis (including lymph node metastases and distant metastases), gastric cancer samples were divided into metastasis and non-metastasis groups. Based on the degree of invasion, gastric cancer specimens were divided into T1 + T2 and T3 + T4 groups. The relative expression of LOX was detected using Western blot. The formation of VM was measured by double staining with CD34 and Periodic acid-Schiff (PAS) in gastric cancer tissue slices, and the correlation between LOX and VM was analyzed with Pearson's correlation analysis. Gastric cancer cell line BGC-803 was treated with LOX, β-aminopropionitrile (BAPN, an inhibitor of LOX), and AG1295 or AG1296 (inhibitors of the platelet-derived growth factor receptor). The formation of VM was then measured using PAS staining. The expression of platelet-derived growth factor receptor (PDGFR)α and PDGFRβ in gastric cancer cells was detected by Western blot. Results: In gastric cancer samples, the level of LOX was higher in the metastasis group than in the non-metastasis group (P < 0.05) and in the T3 + T4 group than in the T1 + T2 group (P < 0.05). VM formation was greater in the T3+T4 group than in the T1+T2 group (P < 0.05) and in the metastasis group than in the non-metastasis group (P < 0.05). The expression level of LOX was positively correlated with VM formation (P < 0.01). In gastric cancer cells, LOX concentration was positively correlated with the degree of VM, and BAPN concentration was negatively correlated with the degree of VM (P <0.05). PDGFR levels in the T3+T4 and metastasis groups were relatively higher (P <0.01) and positively correlated with LOX levels in gastric cancer specimens (P < 0.01). The relative expression of PDGFRα and PDGFRβ in gastric cancer cells was up-regulated with increasing LOX and downregulated with increasing BAPN (P < 0.05). With inhibition of PDGFRα and PDGFRβ using AG1295 or AG1296, VM formation in gastric cancer cells decreased (P <0.05), but the number of VM structures increased while LOX was added (P < 0.05). Conclusion: LOX partially promotes the formation of VM in gastric cancer through the PDGF-PDGFR signaling pathway.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, Ningxia, P.R. China
| | - Bojian Sun
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, Ningxia, P.R. China
- Department of Rheumatology and Immunology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Yuan Lin
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, Ningxia, P.R. China
| | - Hong Deng
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, Ningxia, P.R. China
| | - Xinyi Wang
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, Ningxia, P.R. China
| | - Chuanhao Xu
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, Ningxia, P.R. China
| | - Kaibo Wang
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, Ningxia, P.R. China
| | - Nan Yu
- Department of Rheumatology and Immunology, The General Hospital of Ningxia Medical University, Ningxia 750004, P.R. China
| | - Rongqing Liu
- Department of Rheumatology and Immunology, The General Hospital of Ningxia Medical University, Ningxia 750004, P.R. China
| | - Mei Han
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, Ningxia, P.R. China
| |
Collapse
|
6
|
Zhao Z, Wang Q, Zhao F, Ma J, Sui X, Choe HC, Chen P, Gao X, Zhang L. Single-cell and transcriptomic analyses reveal the influence of diabetes on ovarian cancer. BMC Genomics 2024; 25:1. [PMID: 38166541 PMCID: PMC10759538 DOI: 10.1186/s12864-023-09893-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/11/2023] [Indexed: 01/04/2024] Open
Abstract
BACKGROUND There has been a significant surge in the global prevalence of diabetes mellitus (DM), which increases the susceptibility of individuals to ovarian cancer (OC). However, the relationship between DM and OC remains largely unexplored. The objective of this study is to provide preliminary insights into the shared molecular regulatory mechanisms and potential biomarkers between DM and OC. METHODS Multiple datasets from the GEO database were utilized for bioinformatics analysis. Single cell datasets from the GEO database were analysed. Subsequently, immune cell infiltration analysis was performed on mRNA expression data. The intersection of these datasets yielded a set of common genes associated with both OC and DM. Using these overlapping genes and Cytoscape, a protein‒protein interaction (PPI) network was constructed, and 10 core targets were selected. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were then conducted on these core targets. Additionally, advanced bioinformatics analyses were conducted to construct a TF-mRNA-miRNA coregulatory network based on identified core targets. Furthermore, immunohistochemistry staining (IHC) and real-time quantitative PCR (RT-qPCR) were employed for the validation of the expression and biological functions of core proteins, including HSPAA1, HSPA8, SOD1, and transcription factors SREBF2 and GTAT2, in ovarian tumors. RESULTS The immune cell infiltration analysis based on mRNA expression data for both DM and OC, as well as analysis using single-cell datasets, reveals significant differences in mononuclear cell levels. By intersecting the single-cell datasets, a total of 119 targets related to mononuclear cells in both OC and DM were identified. PPI network analysis further identified 10 hub genesincludingHSP90AA1, HSPA8, SNRPD2, UBA52, SOD1, RPL13A, RPSA, ITGAM, PPP1CC, and PSMA5, as potential targets of OC and DM. Enrichment analysis indicated that these genes are primarily associated with neutrophil degranulation, GDP-dissociation inhibitor activity, and the IL-17 signaling pathway, suggesting their involvement in the regulation of the tumor microenvironment. Furthermore, the TF-gene and miRNA-gene regulatory networks were validated using NetworkAnalyst. The identified TFs included SREBF2, GATA2, and SRF, while the miRNAs included miR-320a, miR-378a-3p, and miR-26a-5p. Simultaneously, IHC and RT-qPCR reveal differential expression of core targets in ovarian tumors after the onset of diabetes. RT-qPCR further revealed that SREBF2 and GATA2 may influence the expression of core proteins, including HSP90AA1, HSPA8, and SOD1. CONCLUSION This study revealed the shared gene interaction network between OC and DM and predicted the TFs and miRNAs associated with core genes in monocytes. Our research findings contribute to identifying potential biological mechanisms underlying the relationship between OC and DM.
Collapse
Affiliation(s)
- Zhihao Zhao
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Qilin Wang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Fang Zhao
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Junnan Ma
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xue Sui
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Hyok Chol Choe
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Department of Clinical Medicine, Sinuiju Medical University, Sinuiju, Democratic People's Republic of Korea
| | - Peng Chen
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xue Gao
- Department of Pathology, the First Hospital of Dalian Medical University, Dalian, Liaoning Province, 116027, China.
| | - Lin Zhang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China.
| |
Collapse
|
7
|
Tadić V, Zhang W, Brozovic A. The high-grade serous ovarian cancer metastasis and chemoresistance in 3D models. Biochim Biophys Acta Rev Cancer 2024; 1879:189052. [PMID: 38097143 DOI: 10.1016/j.bbcan.2023.189052] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most frequent and aggressive type of epithelial ovarian cancer, with high recurrence rate and chemoresistance being the main issues in its clinical management. HGSOC is specifically challenging due to the metastatic dissemination via spheroids in the ascitic fluid. The HGSOC spheroids represent the invasive and chemoresistant cellular fraction, which is impossible to investigate in conventional two-dimensional (2D) monolayer cell cultures lacking critical cell-to-cell and cell-extracellular matrix interactions. Three-dimensional (3D) HGSOC cultures, where cells aggregate and exhibit relevant interactions, offer a promising in vitro model of peritoneal metastasis and multicellular drug resistance. This review summarizes recent studies of HGSOC in 3D culture conditions and highlights the role of multicellular HGSOC spheroids and ascitic environment in HGSOC metastasis and chemoresistance.
Collapse
Affiliation(s)
- Vanja Tadić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, Zagreb HR-10000, Croatia
| | - Wei Zhang
- Department of Engineering Mechanics, Dalian University of Technology, Linggong Road 2, Dalian CN-116024, China
| | - Anamaria Brozovic
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, Zagreb HR-10000, Croatia.
| |
Collapse
|
8
|
Lee B, Chang SJ, Kwon BS, Son JH, Lim MC, Kim YH, Lee SW, Choi CH, Eoh KJ, Lee JY, Suh DH, Kim YB. Clinical guidelines for ovarian cancer: the Korean Society of Gynecologic Oncology guidelines. J Gynecol Oncol 2024; 35:e43. [PMID: 38178704 PMCID: PMC10792210 DOI: 10.3802/jgo.2024.35.e43] [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: 10/25/2023] [Revised: 12/10/2023] [Accepted: 12/13/2023] [Indexed: 01/06/2024] Open
Abstract
Since the latest practice guidelines for ovarian cancer were developed by the Korean Society of Gynecologic Oncology (KSGO) in 2021, many studies have examined the efficacy and safety of various treatments for epithelial ovarian cancer (EOC). Therefore, the need to develop recommendations for EOC treatments has been raised. This study searched the literature using 4 key items and the Population, Intervention, Comparison, and Outcome: the efficacy and safety of poly-ADP ribose polymerase inhibitors in newly diagnosed advanced EOC; the efficacy and safety of intraperitoneal plus intravenous chemotherapy in optimally debulked advanced EOC; the efficacy and safety of secondary cytoreductive surgery in platinum-sensitive recurrent ovarian cancer; and the efficacy and safety of the addition of bevacizumab to platinum-based chemotherapy in first platinum-sensitive recurrent EOC patients who received prior bevacizumab. The evidence for these recommendations, according to each key question, was evaluated using a systematic review and meta-analysis. The committee of ovarian cancer of the KSGO developed updated guidelines for treatments of EOC.
Collapse
Affiliation(s)
- Banghyun Lee
- Department of Obstetrics and Gynecology, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - Suk-Joon Chang
- Department of Obstetrics and Gynecology, Ajou University School of Medicine, Suwon, Korea.
| | - Byung Su Kwon
- Department of Obstetrics and Gynecology, Kyung Hee University Medical Center, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Joo-Hyuk Son
- Department of Obstetrics and Gynecology, Ajou University School of Medicine, Suwon, Korea
| | - Myong Cheol Lim
- Center for Gynecologic Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Yun Hwan Kim
- Department of Obstetrics and Gynecology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Korea
| | - Shin-Wha Lee
- Department of Obstetrics and Gynecology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Chel Hun Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyung Jin Eoh
- Department of Obstetrics and Gynecology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Korea
| | - Dong Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yong Beom Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
9
|
Tang B, Ma W, Lin Y. Emerging applications of anti-angiogenic nanomaterials in oncotherapy. J Control Release 2023; 364:61-78. [PMID: 37871753 DOI: 10.1016/j.jconrel.2023.10.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/08/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
Angiogenesis is the process of generating new blood vessels from pre-existing vasculature. Under normal conditions, this process is delicately controlled by pro-angiogenic and anti-angiogenic factors. Tumor cells can produce plentiful pro-angiogenic molecules promoting pathological angiogenesis for uncontrollable growth. Therefore, anti-angiogenic therapy, which aims to inhibit tumor angiogenesis, has become an attractive approach for oncotherapy. However, classic anti-angiogenic agents have several limitations in clinical use, such as lack of specific targeting, low bioavailability, and poor therapeutic outcomes. Hence, alternative angiogenic inhibitors are highly desired. With the emergence of nanotechnology, various nanomaterials have been designed for anti-angiogenesis purposes, offering promising features like excellent targeting capabilities, reduced side effects, and enhanced therapeutic efficacy. In this review, we describe tumor vascular features, discuss current dilemma of traditional anti-angiogenic medicines in oncotherapy, and underline the potential of nanomaterials in tumor anti-angiogenic therapy. Moreover, we discuss the current challenges of anti-angiogenic cancer treatment. We expect that this summary of anti-angiogenic nanomaterials in oncotherapy will offer valuable insights, facilitating their extensive applications in the future.
Collapse
Affiliation(s)
- Bicai Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
10
|
Zhang C, Qin M. Extracellular vesicles targeting tumor microenvironment in ovarian cancer. Int J Biol Macromol 2023; 252:126300. [PMID: 37573911 DOI: 10.1016/j.ijbiomac.2023.126300] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/17/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
Ovarian cancer (OC) is a prevalent neoplastic condition affecting women. Extracellular vesicles (EVs), nano-sized membrane vesicles, are secreted by various cells in both physiological and pathological states. The profound interplay between EVs and the tumor microenvironment (TME) in ovarian cancer is crucial. In this review, we explores the pivotal role of EVs in facilitating intercellular communication between cancer cells and the TME, emphasizing the potential of EVs as promising diagnostic markers and innovative therapeutic targets for ovarian cancer. The comprehensive analysis outlines the specific mechanisms by which EVs engage in communication with the constituents of the TME, including the modulation of tumor growth through EVs carrying matrix metalloproteinases (MMPs) and EV-mediated inhibition of angiogenesis, among other factors. Additionally, the we discuss the potential clinical applications of EVs that target the TME in ovarian cancer, encompassing the establishment of novel treatment strategies and the identification of novel biomarkers for early detection and prognosis. Finally, this review identifies novel strategies for therapeutic interventions, such as utilizing EVs as carriers for drug delivery and targeting specific EV-mediated signaling pathways. In summary, this manuscript offers valuable insights into the role of EVs in ovarian cancer and highlights the significance of comprehending intercellular communication in the realm of cancer biology.
Collapse
Affiliation(s)
- Chunmei Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, China
| | - Meiying Qin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
11
|
Yu P, Wang Y, Yuan D, Sun Y, Qin S, Li T. Vascular normalization: reshaping the tumor microenvironment and augmenting antitumor immunity for ovarian cancer. Front Immunol 2023; 14:1276694. [PMID: 37936692 PMCID: PMC10626545 DOI: 10.3389/fimmu.2023.1276694] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Ovarian cancer remains a challenging disease with limited treatment options and poor prognosis. The tumor microenvironment (TME) plays a crucial role in tumor growth, progression, and therapy response. One characteristic feature of the TME is the abnormal tumor vasculature, which is associated with inadequate blood perfusion, hypoxia, and immune evasion. Vascular normalization, a therapeutic strategy aiming to rectify the abnormal tumor vasculature, has emerged as a promising approach to reshape the TME, enhance antitumor immunity, and synergize with immunotherapy in ovarian cancer. This review paper provides a comprehensive overview of vascular normalization and its potential implications in ovarian cancer. In this review, we summarize the intricate interplay between anti-angiogenesis and immune modulation, as well as ICI combined with anti-angiogenesis therapy in ovarian cancer. The compelling evidence discussed in this review contributes to the growing body of knowledge supporting the utilization of combination therapy as a promising treatment paradigm for ovarian cancer, paving the way for further clinical development and optimization of this therapeutic approach.
Collapse
Affiliation(s)
- Ping Yu
- Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Yaru Wang
- Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Dahai Yuan
- Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Yunqin Sun
- Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Shuang Qin
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianye Li
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
12
|
Tian X, Si Q, Liu M, Shi J, Zhao R, Xiong Y, Yu L, Cui H, Guan H. Advance in vasculogenic mimicry in ovarian cancer (Review). Oncol Lett 2023; 26:456. [PMID: 37736556 PMCID: PMC10509778 DOI: 10.3892/ol.2023.14043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023] Open
Abstract
Ovarian cancer (OC) is a common and highly prevalent malignant tumor in women, associated with a high mortality rate, easy recurrence and easy metastasis, which is predominantly at an advanced stage when detected in patients. This renders the cancer more difficult to treat, and consequently it is also associated with a low survival rate, being the malignancy with the highest mortality rate among the various gynecological tumors. As an important factor affecting the development and metastasis of OC, understanding the underlying mechanism(s) through which it is formed and developed is crucial in terms of its treatment. At present, the therapeutic methods of angiogenic mimicry for OC remain in the preliminary stages of exploration and have not been applied in actual clinical practice. In the present review, various signaling pathways and factors affecting angiogenic mimicry in OC were described, and the chemical synthetic drugs, natural compound extracts, small-molecule protein antibodies and their associated targets, and so on, that target angiogenic mimicry in the treatment of OC, were discussed. The purpose of this review was to provide new research ideas and potential theoretical support for the discovery of novel therapeutic targets for OC that may be applied in the clinic, with the aim of effectively reducing its metastasis and recurrence rates.
Collapse
Affiliation(s)
- Xinyuan Tian
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| | - Qin Si
- Scientific Research Department, Inner Mongolia Cancer Hospital and Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010020, P.R. China
| | - Menghe Liu
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| | - Jianping Shi
- School of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| | - Rongwei Zhao
- Department of Obstetrics and Gynecology, Inner Mongolia Medical University, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Yang Xiong
- Department of Hepatobiliary Surgery, General Surgery Department of Ordos Central Hospital, Ordos, Inner Mongolia Autonomous Region 017000, P.R. China
| | - Lei Yu
- Department of Pharmacy, Traditional Chinese Medicine Hospital of Inner Mongolia Autonomous Region, Hohhot, Inner Mongolia Autonomous Region 010020, P.R. China
| | - Hongwei Cui
- Scientific Research Department, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Haibin Guan
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| |
Collapse
|
13
|
Lin X, Long S, Yan C, Zou X, Zhang G, Zou J, Wu G. Therapeutic potential of vasculogenic mimicry in urological tumors. Front Oncol 2023; 13:1202656. [PMID: 37810976 PMCID: PMC10551447 DOI: 10.3389/fonc.2023.1202656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
Angiogenesis is an essential process in the growth and metastasis of cancer cells, which can be hampered by an anti-angiogenesis mechanism, thereby delaying the progression of tumors. However, the benefit of this treatment modality could be restricted, as most patients tend to develop acquired resistance during treatment. Vasculogenic mimicry (VM) is regarded as a critical alternative mechanism of tumor angiogenesis, where studies have demonstrated that patients with tumors supplemented with VM generally have a shorter survival period and a poorer prognosis. Inhibiting VM may be an effective therapeutic strategy to prevent cancer progression, which could prove helpful in impeding the limitations of lone use of anti-angiogenic therapy when performed concurrently with other anti-tumor therapies. This review summarizes the mechanism of VM signaling pathways in urological tumors, i.e., prostate cancer, clear cell renal cell carcinoma, and bladder cancer. Furthermore, it also summarizes the potential of VM as a therapeutic strategy for urological tumors.
Collapse
Affiliation(s)
- Xinyu Lin
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Sheng Long
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Congcong Yan
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiaofeng Zou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Guoxi Zhang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junrong Zou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Gengqing Wu
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
14
|
Pu T, Liu Y, Pei Y, Peng J, Wang Z, Du M, Liu Q, Zhong F, Zhang M, Li F, Xu C, Zhang X. NIR-II Fluorescence Imaging for the Detection and Resection of Cancerous Foci and Lymph Nodes in Early-Stage Orthotopic and Advanced-Stage Metastatic Ovarian Cancer Models. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37385963 DOI: 10.1021/acsami.3c04949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
The high mortality rate of ovarian cancer can be primarily attributed to late diagnosis and early lymph node (LN) metastasis. The anatomically deep-located ovaries own intricate anatomical structures and lymphatic drainages that compromise the resolution and sensitivity of near-infrared first-window (NIR-I) fluorescence imaging. Reported NIR-II imaging studies of ovarian cancer focused on late-stage metastasis detection via the intraperitoneal xenograft model. However, given the significant improvement in patient survival associated with early-stage cancer detection, locating tumors that are restricted within the ovary is equally crucial. We obtained the polymer nanoparticles with bright near-infrared-II fluorescence (NIR-II NPs) by nanoprecipitation of DSPE-PEG, one of the ingredients of FDA-approved nanoparticle products, and benzobisthiadiazole, an organic NIR-II dye. The one-step synthesis and safe component lay the groundwork for its clinical translation. Benefiting from the NIR-II emission (∼1060 nm), NIR-II NPs enabled a high signal-to-noise (S/N) ratio (13.4) visualization of early-stage orthotopic ovarian tumors with NIR-II fluorescence imaging for the first time. Imaging with orthotopic xenograft allows a more accurate mimic of human ovarian cancer origin, thereby addressing the dilemma of translating existing nanoprobe preclinical research by providing the nano-bio interactions with early local tumor environments. After PEGylation, the desirable-sized probe (∼80 nm) exhibited high lymphophilicity and relatively extended circulation. NIR-II NPs maintained their accurate detection of orthotopic tumors, tumor-regional LNs, and minuscule (<1 mm) disseminated peritoneal metastases simultaneously (with S/N ratios all above 5) in mice with advanced-stage cancer in real time ∼36 h after systematic delivery. With NIR-II fluorescence guidance, we achieved accurate surgical staging in tumor-bearing mice and complete tumor removal comparable to clinical practice, which provides preclinical data for translating NIR-II fluorescence image-guided surgery.
Collapse
Affiliation(s)
- Tao Pu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Yawei Liu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Yuetian Pei
- Academy for Engineering and Technology, Fudan University, Shanghai 200438, China
| | - Jing Peng
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Zehua Wang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Ming Du
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Qiyu Liu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Fangfang Zhong
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Mingxing Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Fuyou Li
- Department of Chemistry, Fudan University, Shanghai 200438, China
| | - Congjian Xu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Xiaoyan Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| |
Collapse
|
15
|
Fan X, Huang J, Hu B, Zhou J, Chen L. Tumor-expressed B7-H3 promotes vasculogenic mimicry formation rather than angiogenesis in non-small cell lung cancer. J Cancer Res Clin Oncol 2023:10.1007/s00432-023-04790-3. [PMID: 37129607 DOI: 10.1007/s00432-023-04790-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/15/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Vasculogenic mimicry (VM), an alternative microvascular circulation independent of angiogenesis, is formed by aggressive cancer cells. Tumor-expressed B7-H3 has been reported to promote VM formation in hepatocellular carcinoma and modulate angiogenesis in breast cancer and colorectal cancer. However, its effects on VM generation and angiogenesis in non-small cell Lung cancer (NSCLC) remained to be elucidated. METHODS CRISPR/Cas9-mediated B7-H3 knockout (KO) was conducted in NSCLC A549 and H3255 cells. The expression of VM-related proteins, including vascular endothelial (VE)-cadherin and matrix metalloproteinase 14 (MMP14), and the secretion of vascular endothelial growth factor (VEGF) were measured by western blotting and chemiluminescence assay in both B7-H3 KO and mock-edited A549 and H3255 cells. To examine VM formation, a three-dimensional (3D) culture model was used for B7-H3 KO and mock A549 and H3255 cells. For in vivo analysis, xenograft mice models were established using B7-H3 KO and mock-edited A549 cells, and immunohistochemical (CD31) and histochemical (periodic acid-Schiff, PAS) double staining were performed to identify VM and endothelial vessels in tumor tissues. Finally, specific signaling inhibitors were used to analyze B7-H3-induced signaling pathway responsible for VE-cadherin and MMP14 expression and VM generation. RESULTS Higher expression of B7-H3 was associated with a worse prognosis and more advanced T-category in NSCLC. CRISPR/Cas9-mediated B7-H3 KO in A549 and H3255 cells led to decreased expression of VE-cadherin and MMP14; however, the secretion of VEGF by the two cell lines remained unchanged. In the 3D cell culture model, both B7-H3 KO A549 and H3255 cells showed a significant reduction in the formation of capillary-like tubular structures compared to mock-edited cells. In the in vivo xenograft model, mock-edited A549 cells formed excessive PAS+ CD31- VM channels, while B7-H3 KO restrained VM formation in the xenograft tumors. However, no significant differences were found in CD31+ endothelial vessels between xenografts formed by B7-H3 KO and mock-edited A549 cells. Finally, we analyzed the signaling pathway responsible for B7-H3-induced VM formation and found that selective inhibition of the phosphoinositide 3-kinase(PI3K)/protein kinase B (AKT) hyperactivation by LY294002 was associated with decreased expression of MMP14 and VE-cadherin, and in vitro VM formation by both A549 and H3255 cells. CONCLUSIONS Tumor-expressed B7-H3 acts via PI3K/AKT signaling pathway to promote VM formation by NSCLC cells while bears no effects on angiogenesis in NSCLC.
Collapse
Affiliation(s)
- Xingyu Fan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Junfeng Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bingqi Hu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Zhou
- Department of Laboratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liwen Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
16
|
Wang J, Liu Y, Zhang Y, Li X, Fang M, Qian D. Targeting exosomes enveloped EBV-miR-BART1-5p-antagomiRs for NPC therapy through both anti-vasculogenic mimicry and anti-angiogenesis. Cancer Med 2023. [PMID: 37097161 DOI: 10.1002/cam4.5941] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 03/25/2023] [Accepted: 04/01/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a type of head and neck cancer with high incidence in China. The molecular mechanisms of vasculogenic mimicry (VM) and angiogenesis are not fully elucidated in NPC. More specially, it has seldomly been reported that Epstein-Barr virus-encoded miRNA can regulate VM and angiogenesis in NPC. The aim of this study was to investigate the function and molecular mechanism of a targeting exosome system (iRGD-exo-antagomiR) against VM and angiogenesis in NPC, and to provide new approaches for improving the comprehensive treatment of NPC. METHODS Exosomes were isolated by differential ultracentrifugation. Dynamic light scattering, transmission electron microscopy and western blotting were performed to characterize the exosomes. The 3D-Culture assay, tube formation assay, chicken chorioallantoic membrane assay, Matrigel plug assay, mouse xenograft tumor modeling and immunohistochemical staining were applied to evaluate the anti-VM and anti-angiogenic effects of the targeting exosome system in vitro and in vivo. Western blot was performed to detect the changes of downstream regulated networks following interference and recovery of the target gene. RESULTS In vitro or in vivo treatment with iRGD-tagged exosome containing antagomiR-BART1-5p specifically suppressed VM and angiogenesis in NPC. EBV-miR-BART1-5p promoted VM and angiogenesis in vitro and in vivo by regulating VEGF, PI3K, Akt, mTOR and HIF1-α in a Spry2-dependent manner. CONCLUSIONS Our findings demonstrated that targeting exosomes enveloped EBV-miR-BART1-5p-antagomiRs in a Spry2-dependent manner for NPC therapy through both anti-VM and anti-angiogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- Jianguo Wang
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yan Liu
- Health Management center, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yuanbin Zhang
- Shenzhen Key Laboratory of Viral Oncology, the Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xiaoyang Li
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Min Fang
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Dong Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
17
|
Wang YJ, Tang L, Lu XH, Liu JT, Wang YY, Geng HX, Li XT, An Q. Efficacy of epi-1 modified epirubicin and curcumin encapsulated liposomes targeting-EpCAM in the inhibition of epithelial ovarian cancer cells. J Liposome Res 2022:1-17. [PMID: 36440599 DOI: 10.1080/08982104.2022.2153138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Treatment of epithelial ovarian cancer (EOC) is a challenge because it still leads to unsatisfactory clinical prognosis. This is due to the toxicity and poor targeting of chemotherapeutic agents, as well as metastasis of the tumor. In this study, we designed a targeted liposome with nanostructures to overcome these problems. In the liposomes, epirubicin and curcumin were encapsulated to achieve their synergistic antitumor efficacy, while Epi-1 was modified on the liposomal surface to target epithelial cell adhesion molecule (EpCAM). Epi-1, a macrocyclic peptide, exhibits active targeting for enhanced cellular uptake and potent cytotoxicity against tumor cells. The encapsulation of epirubicin and curcumin synergistically inhibited the formation of neovascularization and vasculogenic mimicry (VM) channels, thereby suppressing tumor metastasis on SKOV3 cells. The dual drug loaded Epi-1-liposomes also induced apoptosis and downregulated metastasis-related proteins for effective antitumor in vitro. In vivo studies showed that dual drug loaded Epi-1-liposomes prolonged circulation time in the blood and increased the selective accumulation of drug at the tumor site. H&E staining and immunohistochemistry with Ki-67 also showed that targeted liposomes elevated antitumor activity. Also, targeted liposomes downregulated angiogenesis-related proteins to inhibit angiogenesis and thus tumor metastasis. In conclusion, the production of dual drug loaded Epi-1-liposomes is an effective strategy for the treatment of EOC.
Collapse
Affiliation(s)
- Yu-Jia Wang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Ling Tang
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Xu-Hong Lu
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ji-Tao Liu
- Technology Research and Development Centre, Yunnan Baiyao Group Health Products Co., Ltd, Kunming, China
| | - Yuan-Yuan Wang
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Hong-Xia Geng
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xue-Tao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Quan An
- Technology Research and Development Centre, Yunnan Baiyao Group Health Products Co., Ltd, Kunming, China
| |
Collapse
|
18
|
Molecular Features, Prognostic Value, and Cancer Immune Interactions of Angiogenesis-Related Genes in Ovarian Cancer. Reprod Sci 2022; 30:1637-1650. [PMID: 36471217 DOI: 10.1007/s43032-022-01123-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022]
Abstract
Angiogenesis is crucial to tumor growth and metastasis; it plays a key role in various cancers development and progression. However, the potential effects of angiogenesis-related genes (ARGs) in ovarian cancer (OC) remain to be further investigated. We discussed the characteristics changes of ARGs in 784 OC samples from genomic and transcriptional levels, as well as their expression patterns based on four distinct datasets. First, 784 OC patients were divided into three molecular subtypes, and the findings indicated that ARG changes were correlated with clinicopathological parameters, prognosis, and immune cell-infiltrating tumor microenvironment (TME). OC patients were subsequently divided into two gene subtypes depending on differentially expressed genes (DEGs) of the abovementioned molecular subtypes. We also established an ARGs-related score (ARGs score) model for evaluating overall survival (OS) and determining the immunological landscape of OC patients, therefore predicting patients' prognosis and therapeutic responses. A lower ARGs' score accompanied by a high mutation frequency implies a higher probability of survival. Furthermore, the ARG score was correlated with the cancer stem cell (CSC) index and chemotherapeutic sensitivity. The significant involvement of ARGs in the tumor-immune-stromal microenvironment, clinicopathological characteristics, and prognosis were established in our systematic investigation of ARGs for OC patients. These discoveries might help us to better understand the role of ARGs in OC, as well as give new insight for predicting the prognosis and providing promising immunotherapy.
Collapse
|
19
|
Wang Y, Zhang L, Bai Y, Wang L, Ma X. Therapeutic implications of the tumor microenvironment in ovarian cancer patients receiving PD-1/PD-L1 therapy. Front Immunol 2022; 13:1036298. [PMID: 36341388 PMCID: PMC9630909 DOI: 10.3389/fimmu.2022.1036298] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 09/27/2022] [Indexed: 06/28/2024] Open
Abstract
Epithelial ovarian cancer (EOC) ranks as the second most common cause of gynecologic cancer death. The conventional treatment for patients with EOC is postoperative therapy along with platinum chemotherapy. However, a more efficient treatment regimen is of great need for these patients diagnosed with advanced disease (FIGO stages III-IV), whose survival is approximately 29%. Immunotherapy seems to be an encouraging therapeutic strategy for EOC. Given the crucial role in the complicated interactions between tumor cells and other cells, the tumor microenvironment (TME) influences the response to immunotherapy. In this review, we discuss feasible strategies for EOC immunotherapy by exploiting the reciprocity of cancer cells and the constituents of the TME.
Collapse
Affiliation(s)
- Yusha Wang
- Division of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Zhang
- Department of Obstetrics and Gynecology, Chengdu First People’s Hospital and Chengdu Integrated Traditional Chinese Medicine (TCM) and Western Medicine Hospital, Chengdu, China
| | - Yun Bai
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Li Wang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Division of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Recouvreux MS, Miao J, Gozo MC, Wu J, Walts AE, Karlan BY, Orsulic S. FOXC2 Promotes Vasculogenic Mimicry in Ovarian Cancer. Cancers (Basel) 2022; 14:4851. [PMID: 36230774 PMCID: PMC9564305 DOI: 10.3390/cancers14194851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
FOXC2 is a forkhead family transcription factor that plays a critical role in specifying mesenchymal cell fate during embryogenesis. FOXC2 expression is associated with increased metastasis and poor survival in various solid malignancies. Using in vitro and in vivo assays in mouse ovarian cancer cell lines, we confirmed the previously reported mechanisms by which FOXC2 could promote cancer growth, metastasis, and drug resistance, including epithelial-mesenchymal transition, stem cell-like differentiation, and resistance to anoikis. In addition, we showed that FOXC2 expression is associated with vasculogenic mimicry in mouse and human ovarian cancers. FOXC2 overexpression increased the ability of human ovarian cancer cells to form vascular-like structures in vitro, while inhibition of FOXC2 had the opposite effect. Thus, we present a novel mechanism by which FOXC2 might contribute to cancer aggressiveness and poor patient survival.
Collapse
Affiliation(s)
- Maria Sol Recouvreux
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jiangyong Miao
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Maricel C. Gozo
- Women’s Cancer Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jingni Wu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sandra Orsulic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
| |
Collapse
|
21
|
Hu H, Ma T, Liu N, Hong H, Yu L, Lyu D, Meng X, Wang B, Jiang X. Immunotherapy checkpoints in ovarian cancer vasculogenic mimicry: Tumor immune microenvironments, and drugs. Int Immunopharmacol 2022; 111:109116. [PMID: 35969899 DOI: 10.1016/j.intimp.2022.109116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 02/09/2023]
Abstract
Vasculogenic mimicry (VM), a vessel-like structure independent of endothelial cells, commonly exists in solid tumors which requires blood vessels to grow. As a special source of blood supply for tumor progression to a more aggressive state, VM has been observed in a variety of human malignant tumors and is tightly associated with tumor proliferation, invasion, metastasis, and poor patient prognosis. So far, various factors, including immune cells and cytokines, were reported to regulate ovarian cancer progression by influencing VM formation. Herein, we review the mechanisms that regulate VM formation in ovarian cancer and the effect of cells, cytokines, and signaling molecules in the tumor microenvironment on VM formation, Furthermore, we summarize the current clinical application of drugs targeting VM formation.
Collapse
Affiliation(s)
- Haitao Hu
- Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, PR China.
| | - Ting Ma
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Nanqi Liu
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Hong Hong
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, PR China.
| | - Lujiao Yu
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, PR China.
| | - Dantong Lyu
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Biao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, PR China.
| | - Xuefeng Jiang
- Department of Immunology, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
22
|
Kim I, Choi S, Yoo S, Lee M, Park JW. AURKB, in concert with REST, acts as an oxygen-sensitive epigenetic regulator of the hypoxic induction of MDM2. BMB Rep 2022. [PMID: 35410638 PMCID: PMC9252896 DOI: 10.5483/bmbrep.2022.55.6.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The acute response to hypoxia is mainly driven by hypoxia-inducible factors, but their effects gradually subside with time. Hypoxia-specific histone modifications may be important for the stable maintenance of long-term adaptation to hypoxia. However, little is known about the molecular mechanisms underlying the dynamic alterations of histones under hypoxic conditions. We found that the phosphorylation of histone H3 at Ser-10 (H3S10) was noticeably attenuated after hypoxic challenge, which was mediated by the inhibition of aurora kinase B (AURKB). To understand the role of AURKB in epigenetic regulation, DNA microarray and transcription factor binding site analyses combined with proteomics analysis were performed. Under normoxia, phosphorylated AURKB, in concert with the repressor element-1 silencing transcription factor (REST), phosphorylates H3S10, which allows the AURKB–REST complex to access the MDM2 proto-oncogene. REST then acts as a transcriptional repressor of MDM2 and downregulates its expression. Under hypoxia, AURKB is dephosphorylated and the AURKB–REST complex fails to access MDM2, leading to the upregulation of its expression. In this study, we present a case of hypoxia-specific epigenetic regulation of the oxygen-sensitive AURKB signaling pathway. To better understand the cellular adaptation to hypoxia, it is worthwhile to further investigate the epigenetic regulation of genes under hypoxic conditions.
Collapse
Affiliation(s)
- Iljin Kim
- Department of Pharmacology, Inha University College of Medicine, Incheon 22212, Korea
| | - Sanga Choi
- Department of Pharmacology, Inha University College of Medicine, Incheon 22212, Korea
| | - Seongkyeong Yoo
- Department of Pharmacology, Inha University College of Medicine, Incheon 22212, Korea
| | - Mingyu Lee
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Jong-Wan Park
- Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Korea
- Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
23
|
Li XY, Rao Y, Sun B, Mao XM. Efficacy and Safety of Anlotinib Combined with PD-1 Blockades for Patients with Previously Treated Epithelial Ovarian Cancer: A Retrospective Study. Int J Gen Med 2022; 15:3977-3989. [PMID: 35440872 PMCID: PMC9013415 DOI: 10.2147/ijgm.s352536] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 02/23/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose This study was to investigate the efficacy and safety of anlotinib combined with programmed cell death protein 1 (PD-1) blockades for patients with previously treated advanced epithelial ovarian cancer (EOC). Patients and Methods Present study was designed as a retrospective study, a total of 32 patients with advanced EOC who progressed after at least two lines previously available standard therapy were included in this study. All the patients were administered with anlotinib combined with PD-1 blockades administration. Clinical activity was implemented and analyzed, which was assessed according to the change of target lesion by imaging evidence and all the subjects were followed up regularly. Safety profile were collected and documented during the treatment. Univariate analysis was carried out using log rank test and multivariate analysis were adjusted by Cox regression analysis. Results The best overall response suggested that partial response was noted in 12 patients, stable disease was observed in 14 patients, progressive disease was found in 6 patients. Therefore, the objective response rate (ORR) of the 32 patients was 37.5% (95% CI: 21.1–56.3%), disease control rate (DCR) of the patients was 81.3% (95% CI: 63.6–92.8%). The median follow-up duration of this study was 17.5 months (follow-up range: 0.9–33.5 months). And the median PFS and OS of the 32-patient cohort was 6.8 months (95% CI: 2.64–10.96) and 18.5 months (95% CI: 14.08–22.92), respectively. The most common treatment-related adverse reactions were fatigue (68.8%), nausea and vomiting (56.3%), hypertension (50.0%) and diarrhea (40.6%). Multivariate Cox regression analysis for PFS indicated that ECOG performance status and FIGO stage were independent factors to predict PFS of patients with previously treated EOC. Conclusion Anlotinib combined with PD-1 blockades demonstrated promising efficacy and tolerable safety profile for patients with previously treated advanced EOC preliminarily. The conclusion should be confirmed in more patients with advanced EOC subsequently.
Collapse
Affiliation(s)
- Xiao-Yuan Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, People’s Republic of China
| | - Yang Rao
- Department of Gynecological Oncology, Tianjin Central Obstetrics and Gynecology Hospital, Tianjin, 300199, People’s Republic of China
| | - Bing Sun
- Department of Radiation Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, 100071, People’s Republic of China
| | - Xue-Mei Mao
- Department of Obstetrics and Gynecology, Tianjin Integrated Traditional Chinese and Western Medicine Hospital (Nankai Hospital), Tianjin, 300102, People’s Republic of China
- Correspondence: Xue-Mei Mao; Bing Sun, Tel +86 13820312420; +86 13810193881, Email ;
| |
Collapse
|
24
|
Suh YJ, Lee B, Kim K, Jeong Y, Choi HY, Hwang SO, Kim YB. Bevacizumab versus PARP-inhibitors in women with newly diagnosed ovarian cancer: a network meta-analysis. BMC Cancer 2022; 22:346. [PMID: 35354431 PMCID: PMC8969379 DOI: 10.1186/s12885-022-09455-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In women with newly diagnosed ovarian cancer, bevacizumab and poly (ADP-ribose) polymerase inhibitors (PARPi) exhibit improved progression-free survival (PFS) when administered concurrent with chemotherapy and/or maintenance therapy, but no study has directly compared their effects. Therefore, this study aimed to compare the efficacy and safety of bevacizumab and PARPi in women with newly diagnosed ovarian cancer using a network meta-analysis. METHODS PubMed, Medline, and Embase databases were searched, and five randomized trials assessing PFS in women with newly diagnosed ovarian cancer treated with either bevacizumab, PARPi, or placebo or no additional agent (controls) were identified. PFS was compared in the overall population with ovarian cancer, women with a BRCA1/2 mutation (BRCAm) and women with homologous-recombination deficiency (HRD). Adverse events (grade ≥ 3) were compared in all populations of the included studies. RESULTS PARPi improved PFS significantly more than bevacizumab in women with a BRCAm (HR 0.47; 95% CI 0.36-0.60) and with HRD (HR 0.66; 95% CI 0.50-0.87). However, in the overall population with ovarian cancer, no significant difference in PFS was observed between women treated with PARPi and those treated with bevacizumab. PARPi exhibited the highest surface under the cumulative ranking probabilities value as the most effective treatment for PFS (PARPi vs. bevacizumab: 98% vs. 52% in the overall population with ovarian cancer; 100% vs. 50% in women with BRCAm; 100% vs. 50% in women with HRD). For adverse events, the risk of all treatments was similar. However, PARPi had a higher adverse risk than the control group (relative risk 2.14; 95% CI 1.40-3.26). CONCLUSIONS In women with newly diagnosed ovarian cancer, PARPi might be more effective in terms of PFS compared to bevacizumab. The risk of serious adverse events was similar for PARPi and bevacizumab.
Collapse
Affiliation(s)
- Young Ju Suh
- Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Republic of Korea
| | - Banghyun Lee
- Department of Obstetrics and Gynecology, Inha University hospital, Inha University College of Medicine, 27, Inhang-ro, Sinheung-dong, Jung-gu, Incheon, Republic of Korea.
| | - Kidong Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Yujin Jeong
- Department of Biostatistics, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hwa Yeon Choi
- Department of Obstetrics and Gynecology, Inha University hospital, Inha University College of Medicine, 27, Inhang-ro, Sinheung-dong, Jung-gu, Incheon, Republic of Korea
| | - Sung Ook Hwang
- Department of Obstetrics and Gynecology, Inha University hospital, Inha University College of Medicine, 27, Inhang-ro, Sinheung-dong, Jung-gu, Incheon, Republic of Korea
| | - Yong Beom Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
25
|
Griessinger J, Schwab J, Chen Q, Kühn A, Cotton J, Bowden G, Preibsch H, Reischl G, Quintanilla-Martinez L, Mori H, Dang AN, Kohlhofer U, Aina OH, Borowsky AD, Pichler BJ, Cardiff RD, Schmid AM. Intratumoral in vivo staging of breast cancer by multi-tracer PET and advanced analysis. NPJ Breast Cancer 2022; 8:41. [PMID: 35332139 PMCID: PMC8948294 DOI: 10.1038/s41523-022-00398-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/01/2022] [Indexed: 11/09/2022] Open
Abstract
The staging and local management of breast cancer involves the evaluation of the extent and completeness of excision of both the invasive carcinoma component and also the intraductal component or ductal carcinoma in situ. When both invasive ductal carcinoma and coincident ductal carcinoma in situ are present, assessment of the extent and localization of both components is required for optimal therapeutic planning. We have used a mouse model of breast cancer to evaluate the feasibility of applying molecular imaging to assess the local status of cancers in vivo. Multi-tracer positron emission tomography (PET) and magnetic resonance imaging (MRI) characterize the transition from premalignancy to invasive carcinoma. PET tracers for glucose consumption, membrane synthesis, and neoangiogenesis in combination with a Gaussian mixture model-based analysis reveal image-derived thresholds to separate the different stages within the whole-lesion. Autoradiography, histology, and quantitative image analysis of immunohistochemistry further corroborate our in vivo findings. Finally, clinical data further support our conclusions and demonstrate translational potential. In summary, this preclinical model provides a platform for characterizing multistep tumor progression and provides proof of concept that supports the utilization of advanced protocols for PET/MRI in clinical breast cancer imaging.
Collapse
Affiliation(s)
- Jennifer Griessinger
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Julian Schwab
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany.,Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Qian Chen
- Center for Immunology and Infectious Diseases, University of California, Davis, CA, USA
| | - Anna Kühn
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Jonathan Cotton
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Gregory Bowden
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Heike Preibsch
- Department of Radiology, University Hospital Tuebingen, Tuebingen, Germany
| | - Gerald Reischl
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany.,Cluster of Excellence iFIT(EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| | - Leticia Quintanilla-Martinez
- Cluster of Excellence iFIT(EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany.,Department of Pathology, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Hidetoshi Mori
- Center for Immunology and Infectious Diseases, University of California, Davis, CA, USA
| | - An Nguyen Dang
- Center for Immunology and Infectious Diseases, University of California, Davis, CA, USA
| | - Ursula Kohlhofer
- Department of Pathology, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Olulanu H Aina
- Center for Immunology and Infectious Diseases, University of California, Davis, CA, USA.,Janssen Pharmaceutical, Spring House, PA, USA
| | - Alexander D Borowsky
- Center for Immunology and Infectious Diseases, University of California, Davis, CA, USA
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany.,Cluster of Excellence iFIT(EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany.,German Cancer Consortium (DKTK), Partner Site Tuebingen; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robert D Cardiff
- Center for Immunology and Infectious Diseases, University of California, Davis, CA, USA
| | - Andreas M Schmid
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany. .,Cluster of Excellence iFIT(EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
26
|
Wang M, Ren S, Bi Z, Zhang L, Cui M, Sun R, Bao J, Gao D, Yang B, Li X, Li M, Xiao T, Zhou H, Yang C. Myricetin reverses epithelial–endothelial transition and inhibits vasculogenic mimicry and angiogenesis of hepatocellular carcinoma by directly targeting
PAR1. Phytother Res 2022; 36:1807-1821. [DOI: 10.1002/ptr.7427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Ming Wang
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
| | - Shanfa Ren
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Zhun Bi
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
| | - Liang Zhang
- Department of Thoracic Surgery Tianjin First Central Hospital, Nankai University Tianjin People's Republic of China
| | - Mengqi Cui
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Ronghao Sun
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
| | - Jiali Bao
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Dandi Gao
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Bo Yang
- Department of Thoracic Surgery Tianjin First Central Hospital, Nankai University Tianjin People's Republic of China
| | - Xiaoping Li
- Department of Thoracic Surgery Tianjin First Central Hospital, Nankai University Tianjin People's Republic of China
| | - Mingjiang Li
- Department of Thoracic Surgery Tianjin First Central Hospital, Nankai University Tianjin People's Republic of China
| | - Ting Xiao
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Hong‐gang Zhou
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Cheng Yang
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| |
Collapse
|
27
|
CTCFL regulates the PI3K-Akt pathway and it is a target for personalized ovarian cancer therapy. NPJ Syst Biol Appl 2022; 8:5. [PMID: 35132075 PMCID: PMC8821627 DOI: 10.1038/s41540-022-00214-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 01/05/2022] [Indexed: 12/04/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSC) is the most lethal gynecologic malignancy due to the lack of reliable biomarkers, effective treatment, and chemoresistance. Improving the diagnosis and the development of targeted therapies is still needed. The molecular pathomechanisms driving HGSC progression are not fully understood though crucial for effective diagnosis and identification of novel targeted therapy options. The oncogene CTCFL (BORIS), the paralog of CTCF, is a transcriptional factor highly expressed in ovarian cancer (but in rarely any other tissue in females) with cancer-specific characteristics and therapeutic potential. In this work, we seek to understand the regulatory functions of CTCFL to unravel new target genes with clinical relevance. We used in vitro models to evaluate the transcriptional changes due to the presence of CTCFL, followed by a selection of gene candidates using de novo network enrichment analysis. The resulting mechanistic candidates were further assessed regarding their prognostic potential and druggability. We show that CTCFL-driven genes are involved in cytoplasmic membrane functions; in particular, the PI3K-Akt initiators EGFR1 and VEGFA, as well as ITGB3 and ITGB6 are potential drug targets. Finally, we identified the CTCFL targets ACTBL2, MALT1 and PCDH7 as mechanistic biomarkers to predict survival in HGSC. Finally, we elucidated the value of CTCFL in combination with its targets as a prognostic marker profile for HGSC progression and as putative drug targets.
Collapse
|
28
|
Therapeutic strategies to overcome cisplatin resistance in ovarian cancer. Eur J Med Chem 2022; 232:114205. [DOI: 10.1016/j.ejmech.2022.114205] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 12/15/2022]
|
29
|
Zhuang L, Ming X, Liu J, Jia C, Jin Y, Wang J, Shi Q, Wu R, Jin L, Du L. Comparison of lymphatic contrast-enhanced ultrasound and intravenous contrast-enhanced ultrasound in the preoperative diagnosis of axillary sentinel lymph node metastasis in patients with breast cancer. Br J Radiol 2022; 95:20210897. [PMID: 34797694 PMCID: PMC8822558 DOI: 10.1259/bjr.20210897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVES This study aimed to compare diagnostic efficiency for axillary sentinel lymph node (SLN) metastasis between lymphatic contrast-enhanced ultrasound (LCEUS) and intravenous contrast-enhanced ultrasound (ICEUS) in patients with breast cancer. We also examined whether adding ICEUS to LCEUS could improve the diagnostic accuracy of LCEUS. METHODS Sixty-nine patients with breast cancer were recruited preoperatively. All patients underwent LCEUS followed by ICEUS, and the enhancement pattern of one SLN was analysed for each patient. The targeted SLN was marked with wire and excised during surgery. The imaging diagnosis was compared with the histopathological result. Diagnostic efficiency was compared among LCEUS, ICEUS, and the combination of LCEUS and ICEUS. RESULTS The sensitivity values for LCEUS, ICEUS, and the combination of LCEUS and ICEUS were 86.2%, 82.6% and 93.1%, respectively. Specificity values for the three methods were 95.0%, 92.5% and 87.5%, respectively. Accuracy values for the three methods were 91.3%, 88.4% and 89.9%, respectively. The area under the receiver operating characteristic (ROC) curve for LCEUS was 0.906, and there was no significant difference among LCEUS, ICEUS, and the combination of LCEUS and ICEUS (p = 0.752). CONCLUSIONS LCEUS may represent an accurate method for predicting SLN metastasis preoperatively. Our findings suggest that adding ICEUS to LCEUS for SLN evaluation in patients with breast cancer is unnecessary. ADVANCES IN KNOWLEDGE This is the first study in which both LCEUS and ICEUS were performed for the same lymph node and the first to compare the diagnostic efficiency of LCEUS, ICEUS, and the combination of LCEUS + ICEUS.
Collapse
Affiliation(s)
- Lingling Zhuang
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xu Ming
- Department of Breast-Thyroid-Vascular Surgery, Shanghai General Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Breast-Thyroid-Vascular Surgery, Shanghai General Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, China
| | - Chao Jia
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yubiao Jin
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jing Wang
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Qiusheng Shi
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Rong Wu
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lifang Jin
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lianfang Du
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Han DS, Lee EO. Sp1 Plays a Key Role in Vasculogenic Mimicry of Human Prostate Cancer Cells. Int J Mol Sci 2022; 23:1321. [PMID: 35163245 PMCID: PMC8835864 DOI: 10.3390/ijms23031321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 12/16/2022] Open
Abstract
Sp1 transcription factor regulates genes involved in various phenomena of tumor progression. Vasculogenic mimicry (VM) is the alternative neovascularization by aggressive tumor cells. However, there is no evidence of the relationship between Sp1 and VM. This study investigated whether and how Sp1 plays a crucial role in the process of VM in human prostate cancer (PCa) cell lines, PC-3 and DU145. A cell viability assay and three-dimensional culture VM tube formation assay were performed. Protein and mRNA expression levels were detected by Western blot and reverse transcriptase-polymerase chain reaction, respectively. The nuclear twist expression was observed by immunofluorescence assay. A co-immunoprecipitation assay was performed. Mithramycin A (MiA) and Sp1 siRNA significantly decreased serum-induced VM, whereas Sp1 overexpression caused a significant induction of VM. Serum-upregulated vascular endothelial cadherin (VE-cadherin) protein and mRNA expression levels were decreased after MiA treatment or Sp1 silencing. The protein expression and the nuclear localization of twist were increased by serum, which was effectively inhibited after MiA treatment or Sp1 silencing. The interaction between Sp1 and twist was reduced by MiA. On the contrary, Sp1 overexpression enhanced VE-cadherin and twist expressions. Serum phosphorylated AKT and raised matrix metalloproteinase-2 (MMP-2) and laminin subunit 5 gamma-2 (LAMC2) expressions. MiA or Sp1 silencing impaired these effects. However, Sp1 overexpression upregulated phosphor-AKT, MMP-2 and LAMC2 expressions. Serum-upregulated Sp1 was significantly reduced by an AKT inhibitor, wortmannin. These results demonstrate that Sp1 mediates VM formation through interacting with the twist/VE-cadherin/AKT pathway in human PCa cells.
Collapse
Affiliation(s)
- Deok-Soo Han
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - Eun-Ok Lee
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
- Department of Cancer Preventive Material Development, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| |
Collapse
|
31
|
Yuan Y, Geng B, Xu X, Zhao H, Bai J, Dou Z, Jia S, Yu X, Luo W. Dual VEGF/PDGF knockdown suppresses vasculogenic mimicry formation in choroidal melanoma cells via the Wnt5a/β-catenin/AKT signaling pathway. Acta Histochem 2022; 124:151842. [PMID: 34995928 DOI: 10.1016/j.acthis.2021.151842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVE This study aimed to explore the effects of knocking down both vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF) on vasculogenic mimicry (VM) formation in choroidal melanoma (CM) cells. METHODS Cell counting Kit (CCK)-8, monoclonal formation, wound healing, transwell and flow cytometry assays were used to observe the cell effects in CM cell line, ocular choroidal melanoma-1 cells (OCM-1) with respect to proliferation, migration, invasion and apoptosis. Three-dimensional (3D) cultures were also used to characterize VM tube structural effects in OCM-1 cells and western blotting was used to characterize protein expression changes in VM-related markers. RESULTS Dual VEGF/PDGF knockdown suppressed cell proliferation, migration and invasion, but promoted cell apoptosis. It also reduced VM tube structures in OCM-1 cells. VM associated markers including, VE-cadherin, EphA2 and MT1-MMP were also down-regulated in OCM-1 cells. Similarly, Wnt5a, β-catenin and phosphorylated-AKT levels were also down-regulated. Western blotting and 3D cultures further demonstrated that combined Wnt5a silencing with dual VEGF/PDGF knockdown significantly decreased VE-cadherin and EphA2 levels and reduced VM tube structures in OCM-1 cells. CONCLUSIONS Dual VEGF/PDGF knockdown suppressed cell growth and metastasis in OCM-1 cells, and blocked the Wnt5a/β-catenin/AKT signaling pathway thereby inhibiting VM formation.
Collapse
|
32
|
Salinas-Vera YM, Gallardo-Rincón D, Ruíz-García E, Marchat LA, Valdés J, Vázquez-Calzada C, López-Camarillo C. A Three-Dimensional Culture-Based Assay to Detect Early Stages of Vasculogenic Mimicry in Ovarian Cancer Cells. Methods Mol Biol 2022; 2514:53-60. [PMID: 35771418 DOI: 10.1007/978-1-0716-2403-6_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Vasculogenic mimicry is a cellular mechanism in which tumor cells grow and align forming complex three-dimensional (3D) channel-like structures in a hypoxic microenvironment. This phenomenon represents a novel oxygen, nutrient, and blood supply, in a similar way as occurs in classic angiogenesis. Vasculogenic mimicry has been described in numerous clinical tumors including breast, prostate, lung, and ovarian cancers where it is associated with poor prognosis; thus, it is considered as a hallmark of highly aggressive and metastatic tumors. Here, we describe a simple method to model the in vitro formation of three-dimensional cellular networks over Matrigel in SKOV3 ovarian cancer cells representing the early stages of vasculogenic mimicry.
Collapse
Affiliation(s)
| | - Dolores Gallardo-Rincón
- Laboratory of Translational Medicine and Department of Gastrointestinal Tumors, National Cancer Institute, Ciudad de México, Mexico
| | - Erika Ruíz-García
- Laboratory of Translational Medicine and Department of Gastrointestinal Tumors, National Cancer Institute, Ciudad de México, Mexico
| | - Laurence A Marchat
- Program in Molecular Biomedicine and Biotechnology Network, National Polytechnic Institute, Ciudad de México, Mexico
| | - Jesús Valdés
- Department of Biochemistry, CINVESTAV-IPN, Ciudad de México, Mexico
| | - Carlos Vázquez-Calzada
- Department of Infectomics and Molecular Pathogenesis, CINVESTAV-IPN, Ciudad de México, Mexico
| | | |
Collapse
|
33
|
Vera DB, Fredes AN, Garrido MP, Romero C. Role of Mitochondria in Interplay between NGF/TRKA, miR-145 and Possible Therapeutic Strategies for Epithelial Ovarian Cancer. LIFE (BASEL, SWITZERLAND) 2021; 12:life12010008. [PMID: 35054401 PMCID: PMC8779980 DOI: 10.3390/life12010008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/20/2022]
Abstract
Ovarian cancer is the most lethal gynecological neoplasm, and epithelial ovarian cancer (EOC) accounts for 90% of ovarian malignancies. The 5-year survival is less than 45%, and, unlike other types of cancer, the proportion of women who die from this disease has not improved in recent decades. Nerve growth factor (NGF) and tropomyosin kinase A (TRKA), its high-affinity receptor, play a crucial role in pathogenesis through cell proliferation, angiogenesis, invasion, and migration. NGF/TRKA increase their expression during the progression of EOC by upregulation of oncogenic proteins as vascular endothelial growth factor (VEGF) and c-Myc. Otherwise, the expression of most oncoproteins is regulated by microRNAs (miRs). Our laboratory group reported that the tumoral effect of NGF/TRKA depends on the regulation of miR-145 levels in EOC. Currently, mitochondria have been proposed as new therapeutic targets to activate the apoptotic pathway in the cancer cell. The mitochondria are involved in a myriad of functions as energy production, redox control, homeostasis of Ca+2, and cell death. We demonstrated that NGF stimulation produces an augment in the Bcl-2/BAX ratio, which supports the anti-apoptotic effects of NGF in EOC cells. The review aimed to discuss the role of mitochondria in the interplay between NGF/TRKA and miR-145 and possible therapeutic strategies that may decrease mortality due to EOC.
Collapse
Affiliation(s)
- Daniela B. Vera
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital University of Chile, Santiago 8380456, Chile; (D.B.V.); (A.N.F.)
| | - Allison N. Fredes
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital University of Chile, Santiago 8380456, Chile; (D.B.V.); (A.N.F.)
| | - Maritza P. Garrido
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital University of Chile, Santiago 8380456, Chile; (D.B.V.); (A.N.F.)
- Obstetrics and Gynecology Departament, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Correspondence: (M.P.G.); (C.R.)
| | - Carmen Romero
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital University of Chile, Santiago 8380456, Chile; (D.B.V.); (A.N.F.)
- Obstetrics and Gynecology Departament, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Correspondence: (M.P.G.); (C.R.)
| |
Collapse
|
34
|
Xu Q, Liu Z, Zhu ZQ, Fan Y, Chen R, Xie XH, Cheng M. Knockdown of growth factor receptor bound protein 7 suppresses angiogenesis by inhibiting the secretion of vascular endothelial growth factor A in ovarian cancer cells. Bioengineered 2021; 12:12179-12190. [PMID: 34783299 PMCID: PMC8809950 DOI: 10.1080/21655979.2021.2005225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Growth factor receptor bound protein 7 (GRB7) plays an important role in regulating the growth and metastasis of ovarian cancer. Angiogenesis is the basis for the growth, invasion, and metastasis of malignant tumors. In the current study, we aimed to determine whether GRB7 plays a role in regulating angiogenesis in ovarian cancer. Immunohistochemistry on tissue microarray showed that GRB7 and platelet endothelial cell adhesion molecule-1 (PECAM-1/CD31) protein expression were positively correlated in ovarian cancer tissues. GRB7 knockdown suppressed vascular endothelial growth factor A (VEGFA) expression and reduced VEGFA secretion. The effects of GRB7-silenced SKOV-3 cells on human umbilical vein endothelial cells (HUVECs) were evaluated using a transwell cell co-culture model, which showed that knockdown of GRB7 in SKOV-3 cells suppressed HUVEC proliferation, migration, invasion, and tube formation. Moreover, knockdown of GRB7 in SKOV-3 cells downregulated the expression of proteins associated with angiogenesis, including vascular endothelial growth factor receptor-2 (VEGFR2), mitogen-activated protein kinase kinase 1 (MAP2K1/MEK1), extracellular signal-regulated kinases 1 and 2 (ERK1/2), notch receptor 1 (NOTCH1), and delta-like canonical Notch ligand 4 (DLL4) in HUVECs. In conclusion, knockdown of GRB7 in ovarian cancer cells is an attractive potential therapeutic target for the suppression of angiogenesis in ovarian cancer. GRB7 may regulate angiogenesis through VEGFA/VEGFR2 signaling and its downstream pathways.
Collapse
Affiliation(s)
- Qiong Xu
- Department of Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zequn Liu
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhi-Qin Zhu
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yue Fan
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Rui Chen
- Department of Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiao-Hui Xie
- Department of Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mi Cheng
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
35
|
The anti-ovarian cancer effect of RPV modified paclitaxel plus schisandra B liposomes in SK-OV-3 cells and tumor-bearing mice. Life Sci 2021; 285:120013. [PMID: 34614418 DOI: 10.1016/j.lfs.2021.120013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/23/2021] [Accepted: 09/29/2021] [Indexed: 02/07/2023]
Abstract
AIMS Due to poor targeting ability of anti-tumor drugs and self-adaptation of tumors, the chemotherapy of ovarian cancer is still poorly effective. In recent years, the treatment of tumor with nano-targeted agents has become a potential research focus. In this study, a new type of short cell-penetrating peptide RPV-modified paclitaxel plus schisandrin B liposomes were constructed to disrupt VM channels, angiogenesis, proliferation and migration for the treatment of ovarian cancer. MATERIALS AND METHODS In this study, clone assay, TUNEL, Transwell, wound-healing, CAM and mimics assay were used to detect the effects of RPV-modified liposomes on ovarian cancer SK-OV-3 cells before and after treatment. HE-staining, immunofluorescence and ELISA were used to further detect the expression of tumor-related proteins. KEY FINDINGS RPV-modified paclitaxel plus schisandrin B liposomes can inhibit angiogenesis, VM channel formation, invasion and proliferation of ovarian SK-OV-3 cells. In vitro and in vivo studies showed that tumor-related protein expression was down-regulated. Modification of RPV can prolong the retention time of liposome in vivo and accumulate in the tumor site, increasing the anti-tumor efficacy. SIGNIFICANCE The RPV-modified paclitaxel plus schisandrin B liposomes have good anti-tumor effect, thus may provide a new avenue for the treatment of ovarian cancer.
Collapse
|
36
|
The emerging roles of circular RNAs in vessel co-option and vasculogenic mimicry: clinical insights for anti-angiogenic therapy in cancers. Cancer Metastasis Rev 2021; 41:173-191. [PMID: 34664157 DOI: 10.1007/s10555-021-10000-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/12/2021] [Indexed: 12/20/2022]
Abstract
Unexpected resistance to anti-angiogenic treatment prompted the investigation of non-angiogenic tumor processes. Vessel co-option (VC) and vasculogenic mimicry (VM) are recognized as primary non-angiogenic mechanisms. In VC, cancer cells utilize pre-existing blood vessels for support, whereas in VM, cancer cells channel and provide blood flow to rapidly growing tumors. Both processes have been implicated in the development of tumor and resistance to anti-angiogenic drugs in many tumor types. The morphology, but rare molecular alterations have been investigated in VC and VM. There is a pressing need to better understand the underlying cellular and molecular mechanisms. Here, we review the emerging circular RNA (circRNA)-mediated regulation of non-angiogenic processes, VC and VM.
Collapse
|
37
|
Chen G, Qiu L, Gao J, Wang J, Dang J, Li L, Jin Z, Liu X. Stress Hormones: Emerging Targets in Gynecological Cancers. Front Cell Dev Biol 2021; 9:699487. [PMID: 34307378 PMCID: PMC8299464 DOI: 10.3389/fcell.2021.699487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/09/2021] [Indexed: 01/06/2023] Open
Abstract
In the past decade, several discoveries have documented the existence of innervation in ovarian cancer and cervical cancer. Notably, various neurotransmitters released by the activation of the sympathetic nervous system can promote the proliferation and metastasis of tumor cells and regulate immune cells in the tumor microenvironment. Therefore, a better understanding of the mechanisms involving neurotransmitters in the occurrence and development of gynecological cancers will be beneficial for exploring the feasibility of using inexpensive β-blockers and dopamine agonists in the clinical treatment of gynecological cancers. Additionally, this article provides some new insights into targeting tumor innervation and neurotransmitters in the tumor microenvironment.
Collapse
Affiliation(s)
- Guoqiang Chen
- Department of Obstetrics and Gynecology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lei Qiu
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Jinghai Gao
- Department of Obstetrics and Gynecology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jianhong Dang
- Department of Obstetrics and Gynecology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lingling Li
- Department of Obstetrics and Gynecology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhijun Jin
- Department of Obstetrics and Gynecology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiaojun Liu
- Department of Obstetrics and Gynecology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
38
|
He X, You J, Ding H, Zhang Z, Cui L, Shen X, Bian X, Liu Y, Chen J. Vasculogenic mimicry, a negative indicator for progression free survival of lung adenocarcinoma irrespective of first line treatment and epithelial growth factor receptor mutation status. BMC Cancer 2021; 21:132. [PMID: 33549061 PMCID: PMC7866877 DOI: 10.1186/s12885-021-07863-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
Background Vascular mimicry (VM) was associated with the prognosis of cancers. The aim of the study was to explore the association between VM and anticancer therapy response in patients with lung adenocarcinoma. Methods This was a single-center retrospective study of patients with lung adenocarcinoma between March 1st, 2013, to April 1st, 2019, at the Second People’s Hospital of Taizhou City. All included patients were divided into the VM and no-VM groups according to whether VM was observed or not in the specimen. Vessels with positive PAS and negative CD34 staining were confirmed as VM. The main outcome was progression-free survival (PFS). Results Sixty-six (50.4%) patients were male. Eighty-one patients received chemotherapy as the first-line treatment, and 50 patients received TKIs. Forty-five (34.4%) patients were confirmed with VM. There was no difference regarding the first-line treatment between the VM and no-VM groups (P = 0.285). The 86 patients without VM had a median PFS of 279 (range, 90–1095) days, and 45 patients with VM had a median PFS of 167 (range, 90–369) days (P < 0.001). T stage (hazard ratio (HR) = 1.37, 95% confidence interval (CI): 1.10–1.71), N stage (HR = 1.43, 95%CI: 1.09–1.86), M stage (HR = 2.85, 95%CI: 1.76–4.61), differentiation (HR = 1.85, 95%CI: 1.29–2.65), therapy (HR = 0.32, 95%CI: 0.21–0.49), VM (HR = 2.12, 95%CI: 1.33–3.37), and ECOG (HR = 1.41, 95%CI: 1.09–1.84) were independently associated with PFS. Conclusion The benefits of first-line TKIs for NSCLC with EGFR mutation are possibly better than those of platinum-based regimens in patients without VM, but there is no difference in the benefit of chemotherapy or target therapy for VM-positive NSCLC harboring EGFR mutations.
Collapse
Affiliation(s)
- Xuejun He
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China
| | - Jijun You
- Orthopaedic Department, the Second People's Hospital of Taizhou Affiliated to Medical College of Yangzhou University, Taizhou, China
| | - Haibing Ding
- Orthopaedic Department, the Second People's Hospital of Taizhou Affiliated to Medical College of Yangzhou University, Taizhou, China
| | - Zhisheng Zhang
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China
| | - Lin Cui
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China
| | - Xiaomei Shen
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China
| | - Xiaoxia Bian
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China
| | - Yanqing Liu
- Institute of Medicine, Yangzhou University, No. 88, South Daxue Road, Yangzhou, 225001, China
| | - Jue Chen
- Oncology Department, the Second People's Hospital of Taizhou affiliated to Medical College of Yangzhou University, No. 27, Jiangyan District, Taizhou, 225500, China. .,Institute of Medicine, Yangzhou University, No. 88, South Daxue Road, Yangzhou, 225001, China. .,Respiratory Department, the Second People's Hospital of Taizhou Affiliated to Medical College of Yangzhou University, Taizhou, China.
| |
Collapse
|
39
|
Lim D, Cho JG, Yun E, Lee A, Ryu HY, Lee YJ, Yoon S, Chang W, Lee MS, Kwon BS, Kim J. MicroRNA 34a-AXL Axis Regulates Vasculogenic Mimicry Formation in Breast Cancer Cells. Genes (Basel) 2020; 12:genes12010009. [PMID: 33374832 PMCID: PMC7823537 DOI: 10.3390/genes12010009] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/11/2020] [Accepted: 12/18/2020] [Indexed: 12/31/2022] Open
Abstract
Targeting the tumor vasculature is an attractive strategy for cancer treatment. However, the tumor vasculature is heterogeneous, and the mechanisms involved in the neovascularization of tumors are highly complex. Vasculogenic mimicry (VM) refers to the formation of vessel-like structures by tumor cells, which can contribute to tumor neovascularization, and is closely related to metastasis and a poor prognosis. Here, we report a novel function of AXL receptor tyrosine kinase (AXL) in the regulation of VM formation in breast cancer cells. MDA-MB-231 cells exhibited VM formation on Matrigel cultures, whereas MCF-7 cells did not. Moreover, AXL expression was positively correlated with VM formation. Pharmacological inhibition or AXL knockdown strongly suppressed VM formation in MDA-MB-231 cells, whereas the overexpression of AXL in MCF-7 cells promoted VM formation. In addition, AXL knockdown regulated epithelial–mesenchymal transition (EMT) features, increasing cell invasion and migration in MDA-MB-231 cells. Finally, the overexpression of microRNA-34a (miR-34a), which is a well-described EMT-inhibiting miRNA and targets AXL, inhibited VM formation, migration, and invasion in MDA-MB 231 cells. These results identify a miR-34a–AXL axis that is critical for the regulation of VM formation and may serve as a therapeutic target to inhibit tumor neovascularization.
Collapse
Affiliation(s)
- Dansaem Lim
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (D.L.); (J.G.C.); (E.Y.); (A.L.); (S.Y.); (M.-S.L.)
| | - Jin Gu Cho
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (D.L.); (J.G.C.); (E.Y.); (A.L.); (S.Y.); (M.-S.L.)
| | - Eunsik Yun
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (D.L.); (J.G.C.); (E.Y.); (A.L.); (S.Y.); (M.-S.L.)
| | - Aram Lee
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (D.L.); (J.G.C.); (E.Y.); (A.L.); (S.Y.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Hong-Yeoul Ryu
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of National Sciences, Kyungpook National University, Daegu 41566, Korea;
| | - Young Joo Lee
- Department of Obstetrics and Gynecology, Kyung Hee University Medical Center, 23, Seoul 02447, Korea;
| | - Sukjoon Yoon
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (D.L.); (J.G.C.); (E.Y.); (A.L.); (S.Y.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Korea;
| | - Myeong-Sok Lee
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (D.L.); (J.G.C.); (E.Y.); (A.L.); (S.Y.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Byung Su Kwon
- Department of Obstetrics and Gynecology, Kyung Hee University Medical Center, 23, Seoul 02447, Korea;
- Correspondence: (B.S.K.); (J.K.); Tel.: + 82-2958-8837 (B.S.K.); +82-2710-9553 (J.K.); Fax: +82-2958-8835 (B.S.K.); +82-2-2077-7322 (J.K.)
| | - Jongmin Kim
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (D.L.); (J.G.C.); (E.Y.); (A.L.); (S.Y.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
- Correspondence: (B.S.K.); (J.K.); Tel.: + 82-2958-8837 (B.S.K.); +82-2710-9553 (J.K.); Fax: +82-2958-8835 (B.S.K.); +82-2-2077-7322 (J.K.)
| |
Collapse
|